Do inhaled beta-agonists control cough in URIs or acute bronchitis?

Article Type
Changed
Mon, 01/14/2019 - 13:13
Display Headline
Do inhaled beta-agonists control cough in URIs or acute bronchitis?
EVIDENCE-BASED ANSWER

Patients who receive inhaled beta-agonists for cough due to acute upper respiratory infections (URI) are just as likely to report a productive cough at 7 days compared with patients treated with placebo (strength of recommendation [SOR]: A, based on a systematic review).

One trial, however, showed a reduction in overall cough at 7 days (number needed to treat [NNT]=3, SOR: B, a small randomized controlled trial), and another trial found a reduction in overall symptom score in smokers and those with wheezing on initial exam (SOR: B, based on a small randomized controlled trial).

 

Evidence summary

No studies of inhaled beta-agonists have been conducted with patients who have an explicit diagnosis of acute cough due to URI. While some clinicians feel a distinction between URI and acute bronchitis should be made, there is significant overlap between these diagnoses in clinical practice, as well as in the available studies.

A systematic review looking at beta-agonists for acute bronchitis included the clinical diagnoses of both acute bronchitis and acute cough because a standard definition of bronchitis is lacking.1 Only two trials in this review examined inhaled beta-agonists. When results from these trials were combined for the outcome of productive cough at 7 days, inhaled beta-agonists showed no benefit. However, the authors note that details of the individual trials may help to clarify the effect of inhaled beta-agonists.

One trial, a randomized controlled trial of adult patients with acute bronchitis in 2 community-based family practices, compared 23 patients receiving albuterolin a multidose inhaler (MDI) with 23 patients receiving placebo inhaler.2 Patients were also randomized to receive erythromycin or placebo tablets. Patients with pneumonia or a history of asthma or chronic obstructive pulmonary disease (COPD) were excluded. At 7 days, 61% of patients in the albuterol group reported cough compared with 91% in the control group (P=.02, NNT=3). No statistically significant difference was seen in productive cough or night cough. Smokers responded to inhaled albuterol similarly to nonsmokers. Erythromycin had no effect on cough and side effects were similar among all groups.

The other trial was a randomized controlled trial of 80 adults with cough due to acute respiratory infection; it compared fenoterol aerosol 4 times daily with placebo.3 Inhaled fenoterol is not available in the US but is similar to albuterol. This study showed no difference in cough at 7 days (relative risk [RR]=0.83; 95% confidence interval [CI], 0.52–1.30). In a subgroup analysis, however, smokers and those wheezing on initial exam had lower overall symptom scores when treated with fenoterol.

Recommendations from others

We were unable to find any guidelines on the use of albuterol via MDI for cough from bronchitis or URIs.

CLINICAL COMMENTARY:

Inhaled beta-agonists may aid symptoms; other outcomes may not be improved

Even without a history of lung disease, patients presenting with cough due to acute respiratory illness and with evidence of airflow obstruction (wheezing) appear to receive symptom relief from inhaled beta-agonists. Smokers may be another subgroup who benefit from treatment. However, important patient-oriented outcomes (such as reduced need for over-the-counter medicines, general well being, and return to work) do not improve. If using inhaled albuterol to treat acute cough in practice, one must also consider the financial costs and adverse effects associated with treatment.

References

1. Smucny J, Flynn C, Becker L, Glazier R. Beta2-agonists for acute bronchitis (Cochrane Review). In: The Cochrane Library, Issue 2, 2004. Chichester, UK: John Wiley & Sons, Ltd.

2. Hueston WJ. Albuterol delivered by metered-dose inhaler to treat acute bronchitis. J Fam Pract 1994;39:437-440.

3. Melbye H, Aasebo U, Straume B. Symptomatic effect of inhaled fenoterol in acute bronchitis: a placebo controlled double-blind study. Fam Pract 1991;8:216-222.

Article PDF
Author and Disclosure Information

Mary Maniscalco Stephens, MD, MPH
East Tennessee State University, Johnson City, TN;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Inc, Iowa City, IA

Issue
The Journal of Family Practice - 53(8)
Publications
Topics
Page Number
650-663
Sections
Author and Disclosure Information

Mary Maniscalco Stephens, MD, MPH
East Tennessee State University, Johnson City, TN;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Inc, Iowa City, IA

Author and Disclosure Information

Mary Maniscalco Stephens, MD, MPH
East Tennessee State University, Johnson City, TN;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Inc, Iowa City, IA

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Patients who receive inhaled beta-agonists for cough due to acute upper respiratory infections (URI) are just as likely to report a productive cough at 7 days compared with patients treated with placebo (strength of recommendation [SOR]: A, based on a systematic review).

One trial, however, showed a reduction in overall cough at 7 days (number needed to treat [NNT]=3, SOR: B, a small randomized controlled trial), and another trial found a reduction in overall symptom score in smokers and those with wheezing on initial exam (SOR: B, based on a small randomized controlled trial).

 

Evidence summary

No studies of inhaled beta-agonists have been conducted with patients who have an explicit diagnosis of acute cough due to URI. While some clinicians feel a distinction between URI and acute bronchitis should be made, there is significant overlap between these diagnoses in clinical practice, as well as in the available studies.

A systematic review looking at beta-agonists for acute bronchitis included the clinical diagnoses of both acute bronchitis and acute cough because a standard definition of bronchitis is lacking.1 Only two trials in this review examined inhaled beta-agonists. When results from these trials were combined for the outcome of productive cough at 7 days, inhaled beta-agonists showed no benefit. However, the authors note that details of the individual trials may help to clarify the effect of inhaled beta-agonists.

One trial, a randomized controlled trial of adult patients with acute bronchitis in 2 community-based family practices, compared 23 patients receiving albuterolin a multidose inhaler (MDI) with 23 patients receiving placebo inhaler.2 Patients were also randomized to receive erythromycin or placebo tablets. Patients with pneumonia or a history of asthma or chronic obstructive pulmonary disease (COPD) were excluded. At 7 days, 61% of patients in the albuterol group reported cough compared with 91% in the control group (P=.02, NNT=3). No statistically significant difference was seen in productive cough or night cough. Smokers responded to inhaled albuterol similarly to nonsmokers. Erythromycin had no effect on cough and side effects were similar among all groups.

The other trial was a randomized controlled trial of 80 adults with cough due to acute respiratory infection; it compared fenoterol aerosol 4 times daily with placebo.3 Inhaled fenoterol is not available in the US but is similar to albuterol. This study showed no difference in cough at 7 days (relative risk [RR]=0.83; 95% confidence interval [CI], 0.52–1.30). In a subgroup analysis, however, smokers and those wheezing on initial exam had lower overall symptom scores when treated with fenoterol.

Recommendations from others

We were unable to find any guidelines on the use of albuterol via MDI for cough from bronchitis or URIs.

CLINICAL COMMENTARY:

Inhaled beta-agonists may aid symptoms; other outcomes may not be improved

Even without a history of lung disease, patients presenting with cough due to acute respiratory illness and with evidence of airflow obstruction (wheezing) appear to receive symptom relief from inhaled beta-agonists. Smokers may be another subgroup who benefit from treatment. However, important patient-oriented outcomes (such as reduced need for over-the-counter medicines, general well being, and return to work) do not improve. If using inhaled albuterol to treat acute cough in practice, one must also consider the financial costs and adverse effects associated with treatment.

EVIDENCE-BASED ANSWER

Patients who receive inhaled beta-agonists for cough due to acute upper respiratory infections (URI) are just as likely to report a productive cough at 7 days compared with patients treated with placebo (strength of recommendation [SOR]: A, based on a systematic review).

One trial, however, showed a reduction in overall cough at 7 days (number needed to treat [NNT]=3, SOR: B, a small randomized controlled trial), and another trial found a reduction in overall symptom score in smokers and those with wheezing on initial exam (SOR: B, based on a small randomized controlled trial).

 

Evidence summary

No studies of inhaled beta-agonists have been conducted with patients who have an explicit diagnosis of acute cough due to URI. While some clinicians feel a distinction between URI and acute bronchitis should be made, there is significant overlap between these diagnoses in clinical practice, as well as in the available studies.

A systematic review looking at beta-agonists for acute bronchitis included the clinical diagnoses of both acute bronchitis and acute cough because a standard definition of bronchitis is lacking.1 Only two trials in this review examined inhaled beta-agonists. When results from these trials were combined for the outcome of productive cough at 7 days, inhaled beta-agonists showed no benefit. However, the authors note that details of the individual trials may help to clarify the effect of inhaled beta-agonists.

One trial, a randomized controlled trial of adult patients with acute bronchitis in 2 community-based family practices, compared 23 patients receiving albuterolin a multidose inhaler (MDI) with 23 patients receiving placebo inhaler.2 Patients were also randomized to receive erythromycin or placebo tablets. Patients with pneumonia or a history of asthma or chronic obstructive pulmonary disease (COPD) were excluded. At 7 days, 61% of patients in the albuterol group reported cough compared with 91% in the control group (P=.02, NNT=3). No statistically significant difference was seen in productive cough or night cough. Smokers responded to inhaled albuterol similarly to nonsmokers. Erythromycin had no effect on cough and side effects were similar among all groups.

The other trial was a randomized controlled trial of 80 adults with cough due to acute respiratory infection; it compared fenoterol aerosol 4 times daily with placebo.3 Inhaled fenoterol is not available in the US but is similar to albuterol. This study showed no difference in cough at 7 days (relative risk [RR]=0.83; 95% confidence interval [CI], 0.52–1.30). In a subgroup analysis, however, smokers and those wheezing on initial exam had lower overall symptom scores when treated with fenoterol.

Recommendations from others

We were unable to find any guidelines on the use of albuterol via MDI for cough from bronchitis or URIs.

CLINICAL COMMENTARY:

Inhaled beta-agonists may aid symptoms; other outcomes may not be improved

Even without a history of lung disease, patients presenting with cough due to acute respiratory illness and with evidence of airflow obstruction (wheezing) appear to receive symptom relief from inhaled beta-agonists. Smokers may be another subgroup who benefit from treatment. However, important patient-oriented outcomes (such as reduced need for over-the-counter medicines, general well being, and return to work) do not improve. If using inhaled albuterol to treat acute cough in practice, one must also consider the financial costs and adverse effects associated with treatment.

References

1. Smucny J, Flynn C, Becker L, Glazier R. Beta2-agonists for acute bronchitis (Cochrane Review). In: The Cochrane Library, Issue 2, 2004. Chichester, UK: John Wiley & Sons, Ltd.

2. Hueston WJ. Albuterol delivered by metered-dose inhaler to treat acute bronchitis. J Fam Pract 1994;39:437-440.

3. Melbye H, Aasebo U, Straume B. Symptomatic effect of inhaled fenoterol in acute bronchitis: a placebo controlled double-blind study. Fam Pract 1991;8:216-222.

References

1. Smucny J, Flynn C, Becker L, Glazier R. Beta2-agonists for acute bronchitis (Cochrane Review). In: The Cochrane Library, Issue 2, 2004. Chichester, UK: John Wiley & Sons, Ltd.

2. Hueston WJ. Albuterol delivered by metered-dose inhaler to treat acute bronchitis. J Fam Pract 1994;39:437-440.

3. Melbye H, Aasebo U, Straume B. Symptomatic effect of inhaled fenoterol in acute bronchitis: a placebo controlled double-blind study. Fam Pract 1991;8:216-222.

Issue
The Journal of Family Practice - 53(8)
Issue
The Journal of Family Practice - 53(8)
Page Number
650-663
Page Number
650-663
Publications
Publications
Topics
Article Type
Display Headline
Do inhaled beta-agonists control cough in URIs or acute bronchitis?
Display Headline
Do inhaled beta-agonists control cough in URIs or acute bronchitis?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

Does lowering diastolic BP to less than 90 mm Hg decrease cardiovascular risk?

Article Type
Changed
Mon, 01/14/2019 - 13:11
Display Headline
Does lowering diastolic BP to less than 90 mm Hg decrease cardiovascular risk?
EVIDENCE-BASED ANSWER

Although lowering diastolic blood pressure (DBP) is associated with reduced cardiovascular events, systolic blood pressure (SBP) is a more robust predictor of cardiovascular risk than DBP and should now be used to diagnose, stage, and treat hypertension.

Lowering diastolic blood pressure (DBP) to <90 mm Hg in hypertensive individuals of all ages decreases the risk of cardiovascular events including myocardial infarction (MI), heart failure, and sudden death (strength of recommendation [SOR]: A, based on systematic review of randomized controlled trials). However, there is no consensus regarding how far to lower DBP. A “J-shaped” increase in cardiovascular risks with DBP <85 mm Hg may apply under certain conditions.

 

Evidence summary

The concept of a continuous graded relationship between DBP and cardiovascular risk is supported by a meta-analysis of 14 randomized clinical trials showing that lowering DBP by 6 mm Hg reduced the risk of coronary heart disease by 14% (95% confidence interval [CI], 4%–22%; P<.01; NNT=200).1 Throughout the range of DBP in study subjects, 70–115 mm Hg, a lower DBP was associated with a lower risk of coronary heart disease.

However, there is concern that lowering DBP too much may actually increase cardiovascular risk. A 10-year observational study showed that in patients with a history of ischemic heart disease, the incidence of fatal MI was lowest when DBP was between 85 to 90 mm Hg and increased with DBP <85 mm Hg, thus demonstrating a J-shaped curve.2

Farnett et al3 derived a summary curve from 13 studies that stratified cardiovascular outcomes by level of achieved blood pressure; the nadir of the curve for ischemic heart disease events occurred at 86 to 89 mm Hg DBP. The risk was independent of type of drug therapy, and more pronounced in study subjects with known cardiovascular disease.

A meta-analysis of 7 randomized controlled trials involving 40,233 hypertensive patients used statistical modeling to determine the shape of the “mortality curve” over a range of DBP categories, defined in 10-mm Hg increments from 65 to 106. The subjects received mainly beta-blockers or thiazide diuretics; controls received placebo or no treatment.4 Both groups demonstrated increased risk for cardiovascular and all-cause death at the lowest DBP levels. Among treated patients, overall death rate was lowest with a DBP in the range of 76 to 85 mm Hg; among controls the nadir was 86 to 95 mm Hg.

The Hypertension Optimal Treatment (HOT) trial5 was specifically designed to determine the optimal target blood pressure for hypertensive patients: 18,790 men and women with DBP 100 to 115 mm Hg were randomly assigned to target DBP groups of <90, <85, or <80 mm Hg. All were treated with felodipine and other agents in a stepped-care protocol; average follow-up was 3.8 years. The lowest incidence of cardiovascular events occurred at a mean DBP of 82.6 mm Hg and fewest cardiovascular deaths at 86.5 mm Hg. Further reductions in DBP neither lowered nor increased cardiovascular risk.

A French cohort study6 followed over 4700 hyper-tensive men for an average of 14 years. These men had their hypertension treated in usual fashion by their own physicians. In this group, SBP was much more accurate than DBP in classifying severity of hypertension and in predicting cardiovascular risk.

Recommendations from others

The Joint National Committee on Prevention, Detection, Evaluation and Treatment of High Blood Pressure (JNC VII)7 and the World Health Organization–International Society of Hyper-tension Guidelines8 state that the relationship between cardiovascular risk and blood pressure is continuous, without a lower threshold. Target blood pressure goals are <140/90 mm Hg in uncomplicated hypertension and <130/80 mm Hg for individuals with diabetes or kidney disease. The National High Blood Pressure Education Program stressed that SBP, not DBP, should become the major criterion for diagnosis and treatment of hypertension.9

CLINICAL COMMENTARY

Emphasize education and focus on systolic blood pressure
Randy Ward, MD
Director, Family Medicine/Psychiatry Residency, Medical College of Wisconsin, Milwaukee

In light of JNC VII, there may be some confusion on the part of patients as to “normal” blood pressure and indications for treatment. In fact, on the first page of the NHLBI web site, “Your Guide to Lowering Blood Pressure,” the statement is made that “normal blood pressure is less than 120 mm Hg systolic and less than 80 mm Hg diastolic.” They later go on to describe the category of prehypertension. It is important to understand the concept and implications of prehypertension, and the “J-shaped” curve in counseling our patients on achieving optimal blood pressure control.

References

1. Collins R, Peto R, MacMahon S, et al. Blood pressure, stroke, and coronary heart disease. Part 2, Short-term reductions in blood pressure: overview of randomised drug trials in their epidemiological context. Lancet 1990;335:827-838.

2. Cruickshank JM, Thorp JM, Zacharias FJ. Benefits and potential harm of lowering high blood pressure. Lancet 1987;1:581-584.

3. Farnett L, Mulrow CD, Linn WD, Lucey CR, Tuley MR. The J-curve phenomenon and the treatment of hypertension. Is there a point beyond which pressure reduction is dangerous? JAMA 1991;265:489-495.

4. Boutitie F, Gueyffier F, Pocock S, Fagard R, Boissel JP. Jshaped relationship between blood pressure and mortality in hypertensive patients: new insights from a meta-analysis of individual-patient data. Ann Intern Med 2002;136:438-448.

5. Hansson L, Zanchetti A, Carruthers SG, et al. Effects of intensive blood pressure lowering and low-dose aspirin in patients treated with hypertension: principal results of the Hypertension Optimal Treatment (HOT) randomised trial. HOT Study Group. Lancet 1998;351:1755-1762.

6. Benetos A, Thomas F, Bean K, Gautier S, Smulyan H, Guize L. Prognostic value of systolic and diastolic blood pressure in treated hypertensive men. Arch Intern Med 2002;162:577-581.

7. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 Report. JAMA 2003;289:2560-2572.

8. 1999 World Health Organization–International Society of Hypertension Guidelines for the Management of Hypertension. Guidelines Subcommittee. J Hypertens 1999;17:151-183.

9. Izzo JL, Jr, Levy D, Black HR. Clinical Advisory Statement. Importance of systolic blood pressure in older Americans. Hypertension 2000;35:1021-1024.

Article PDF
Author and Disclosure Information

Patricia Fontaine, MD, MS
Department of Family Practice and Community Health, University Family Physicians–North Memorial Clinic, Minneapolis, Minn;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Issue
The Journal of Family Practice - 53(2)
Publications
Topics
Page Number
138-156
Sections
Author and Disclosure Information

Patricia Fontaine, MD, MS
Department of Family Practice and Community Health, University Family Physicians–North Memorial Clinic, Minneapolis, Minn;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Author and Disclosure Information

Patricia Fontaine, MD, MS
Department of Family Practice and Community Health, University Family Physicians–North Memorial Clinic, Minneapolis, Minn;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Although lowering diastolic blood pressure (DBP) is associated with reduced cardiovascular events, systolic blood pressure (SBP) is a more robust predictor of cardiovascular risk than DBP and should now be used to diagnose, stage, and treat hypertension.

Lowering diastolic blood pressure (DBP) to <90 mm Hg in hypertensive individuals of all ages decreases the risk of cardiovascular events including myocardial infarction (MI), heart failure, and sudden death (strength of recommendation [SOR]: A, based on systematic review of randomized controlled trials). However, there is no consensus regarding how far to lower DBP. A “J-shaped” increase in cardiovascular risks with DBP <85 mm Hg may apply under certain conditions.

 

Evidence summary

The concept of a continuous graded relationship between DBP and cardiovascular risk is supported by a meta-analysis of 14 randomized clinical trials showing that lowering DBP by 6 mm Hg reduced the risk of coronary heart disease by 14% (95% confidence interval [CI], 4%–22%; P<.01; NNT=200).1 Throughout the range of DBP in study subjects, 70–115 mm Hg, a lower DBP was associated with a lower risk of coronary heart disease.

However, there is concern that lowering DBP too much may actually increase cardiovascular risk. A 10-year observational study showed that in patients with a history of ischemic heart disease, the incidence of fatal MI was lowest when DBP was between 85 to 90 mm Hg and increased with DBP <85 mm Hg, thus demonstrating a J-shaped curve.2

Farnett et al3 derived a summary curve from 13 studies that stratified cardiovascular outcomes by level of achieved blood pressure; the nadir of the curve for ischemic heart disease events occurred at 86 to 89 mm Hg DBP. The risk was independent of type of drug therapy, and more pronounced in study subjects with known cardiovascular disease.

A meta-analysis of 7 randomized controlled trials involving 40,233 hypertensive patients used statistical modeling to determine the shape of the “mortality curve” over a range of DBP categories, defined in 10-mm Hg increments from 65 to 106. The subjects received mainly beta-blockers or thiazide diuretics; controls received placebo or no treatment.4 Both groups demonstrated increased risk for cardiovascular and all-cause death at the lowest DBP levels. Among treated patients, overall death rate was lowest with a DBP in the range of 76 to 85 mm Hg; among controls the nadir was 86 to 95 mm Hg.

The Hypertension Optimal Treatment (HOT) trial5 was specifically designed to determine the optimal target blood pressure for hypertensive patients: 18,790 men and women with DBP 100 to 115 mm Hg were randomly assigned to target DBP groups of <90, <85, or <80 mm Hg. All were treated with felodipine and other agents in a stepped-care protocol; average follow-up was 3.8 years. The lowest incidence of cardiovascular events occurred at a mean DBP of 82.6 mm Hg and fewest cardiovascular deaths at 86.5 mm Hg. Further reductions in DBP neither lowered nor increased cardiovascular risk.

A French cohort study6 followed over 4700 hyper-tensive men for an average of 14 years. These men had their hypertension treated in usual fashion by their own physicians. In this group, SBP was much more accurate than DBP in classifying severity of hypertension and in predicting cardiovascular risk.

Recommendations from others

The Joint National Committee on Prevention, Detection, Evaluation and Treatment of High Blood Pressure (JNC VII)7 and the World Health Organization–International Society of Hyper-tension Guidelines8 state that the relationship between cardiovascular risk and blood pressure is continuous, without a lower threshold. Target blood pressure goals are <140/90 mm Hg in uncomplicated hypertension and <130/80 mm Hg for individuals with diabetes or kidney disease. The National High Blood Pressure Education Program stressed that SBP, not DBP, should become the major criterion for diagnosis and treatment of hypertension.9

CLINICAL COMMENTARY

Emphasize education and focus on systolic blood pressure
Randy Ward, MD
Director, Family Medicine/Psychiatry Residency, Medical College of Wisconsin, Milwaukee

In light of JNC VII, there may be some confusion on the part of patients as to “normal” blood pressure and indications for treatment. In fact, on the first page of the NHLBI web site, “Your Guide to Lowering Blood Pressure,” the statement is made that “normal blood pressure is less than 120 mm Hg systolic and less than 80 mm Hg diastolic.” They later go on to describe the category of prehypertension. It is important to understand the concept and implications of prehypertension, and the “J-shaped” curve in counseling our patients on achieving optimal blood pressure control.

EVIDENCE-BASED ANSWER

Although lowering diastolic blood pressure (DBP) is associated with reduced cardiovascular events, systolic blood pressure (SBP) is a more robust predictor of cardiovascular risk than DBP and should now be used to diagnose, stage, and treat hypertension.

Lowering diastolic blood pressure (DBP) to <90 mm Hg in hypertensive individuals of all ages decreases the risk of cardiovascular events including myocardial infarction (MI), heart failure, and sudden death (strength of recommendation [SOR]: A, based on systematic review of randomized controlled trials). However, there is no consensus regarding how far to lower DBP. A “J-shaped” increase in cardiovascular risks with DBP <85 mm Hg may apply under certain conditions.

 

Evidence summary

The concept of a continuous graded relationship between DBP and cardiovascular risk is supported by a meta-analysis of 14 randomized clinical trials showing that lowering DBP by 6 mm Hg reduced the risk of coronary heart disease by 14% (95% confidence interval [CI], 4%–22%; P<.01; NNT=200).1 Throughout the range of DBP in study subjects, 70–115 mm Hg, a lower DBP was associated with a lower risk of coronary heart disease.

However, there is concern that lowering DBP too much may actually increase cardiovascular risk. A 10-year observational study showed that in patients with a history of ischemic heart disease, the incidence of fatal MI was lowest when DBP was between 85 to 90 mm Hg and increased with DBP <85 mm Hg, thus demonstrating a J-shaped curve.2

Farnett et al3 derived a summary curve from 13 studies that stratified cardiovascular outcomes by level of achieved blood pressure; the nadir of the curve for ischemic heart disease events occurred at 86 to 89 mm Hg DBP. The risk was independent of type of drug therapy, and more pronounced in study subjects with known cardiovascular disease.

A meta-analysis of 7 randomized controlled trials involving 40,233 hypertensive patients used statistical modeling to determine the shape of the “mortality curve” over a range of DBP categories, defined in 10-mm Hg increments from 65 to 106. The subjects received mainly beta-blockers or thiazide diuretics; controls received placebo or no treatment.4 Both groups demonstrated increased risk for cardiovascular and all-cause death at the lowest DBP levels. Among treated patients, overall death rate was lowest with a DBP in the range of 76 to 85 mm Hg; among controls the nadir was 86 to 95 mm Hg.

The Hypertension Optimal Treatment (HOT) trial5 was specifically designed to determine the optimal target blood pressure for hypertensive patients: 18,790 men and women with DBP 100 to 115 mm Hg were randomly assigned to target DBP groups of <90, <85, or <80 mm Hg. All were treated with felodipine and other agents in a stepped-care protocol; average follow-up was 3.8 years. The lowest incidence of cardiovascular events occurred at a mean DBP of 82.6 mm Hg and fewest cardiovascular deaths at 86.5 mm Hg. Further reductions in DBP neither lowered nor increased cardiovascular risk.

A French cohort study6 followed over 4700 hyper-tensive men for an average of 14 years. These men had their hypertension treated in usual fashion by their own physicians. In this group, SBP was much more accurate than DBP in classifying severity of hypertension and in predicting cardiovascular risk.

Recommendations from others

The Joint National Committee on Prevention, Detection, Evaluation and Treatment of High Blood Pressure (JNC VII)7 and the World Health Organization–International Society of Hyper-tension Guidelines8 state that the relationship between cardiovascular risk and blood pressure is continuous, without a lower threshold. Target blood pressure goals are <140/90 mm Hg in uncomplicated hypertension and <130/80 mm Hg for individuals with diabetes or kidney disease. The National High Blood Pressure Education Program stressed that SBP, not DBP, should become the major criterion for diagnosis and treatment of hypertension.9

CLINICAL COMMENTARY

Emphasize education and focus on systolic blood pressure
Randy Ward, MD
Director, Family Medicine/Psychiatry Residency, Medical College of Wisconsin, Milwaukee

In light of JNC VII, there may be some confusion on the part of patients as to “normal” blood pressure and indications for treatment. In fact, on the first page of the NHLBI web site, “Your Guide to Lowering Blood Pressure,” the statement is made that “normal blood pressure is less than 120 mm Hg systolic and less than 80 mm Hg diastolic.” They later go on to describe the category of prehypertension. It is important to understand the concept and implications of prehypertension, and the “J-shaped” curve in counseling our patients on achieving optimal blood pressure control.

References

1. Collins R, Peto R, MacMahon S, et al. Blood pressure, stroke, and coronary heart disease. Part 2, Short-term reductions in blood pressure: overview of randomised drug trials in their epidemiological context. Lancet 1990;335:827-838.

2. Cruickshank JM, Thorp JM, Zacharias FJ. Benefits and potential harm of lowering high blood pressure. Lancet 1987;1:581-584.

3. Farnett L, Mulrow CD, Linn WD, Lucey CR, Tuley MR. The J-curve phenomenon and the treatment of hypertension. Is there a point beyond which pressure reduction is dangerous? JAMA 1991;265:489-495.

4. Boutitie F, Gueyffier F, Pocock S, Fagard R, Boissel JP. Jshaped relationship between blood pressure and mortality in hypertensive patients: new insights from a meta-analysis of individual-patient data. Ann Intern Med 2002;136:438-448.

5. Hansson L, Zanchetti A, Carruthers SG, et al. Effects of intensive blood pressure lowering and low-dose aspirin in patients treated with hypertension: principal results of the Hypertension Optimal Treatment (HOT) randomised trial. HOT Study Group. Lancet 1998;351:1755-1762.

6. Benetos A, Thomas F, Bean K, Gautier S, Smulyan H, Guize L. Prognostic value of systolic and diastolic blood pressure in treated hypertensive men. Arch Intern Med 2002;162:577-581.

7. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 Report. JAMA 2003;289:2560-2572.

8. 1999 World Health Organization–International Society of Hypertension Guidelines for the Management of Hypertension. Guidelines Subcommittee. J Hypertens 1999;17:151-183.

9. Izzo JL, Jr, Levy D, Black HR. Clinical Advisory Statement. Importance of systolic blood pressure in older Americans. Hypertension 2000;35:1021-1024.

References

1. Collins R, Peto R, MacMahon S, et al. Blood pressure, stroke, and coronary heart disease. Part 2, Short-term reductions in blood pressure: overview of randomised drug trials in their epidemiological context. Lancet 1990;335:827-838.

2. Cruickshank JM, Thorp JM, Zacharias FJ. Benefits and potential harm of lowering high blood pressure. Lancet 1987;1:581-584.

3. Farnett L, Mulrow CD, Linn WD, Lucey CR, Tuley MR. The J-curve phenomenon and the treatment of hypertension. Is there a point beyond which pressure reduction is dangerous? JAMA 1991;265:489-495.

4. Boutitie F, Gueyffier F, Pocock S, Fagard R, Boissel JP. Jshaped relationship between blood pressure and mortality in hypertensive patients: new insights from a meta-analysis of individual-patient data. Ann Intern Med 2002;136:438-448.

5. Hansson L, Zanchetti A, Carruthers SG, et al. Effects of intensive blood pressure lowering and low-dose aspirin in patients treated with hypertension: principal results of the Hypertension Optimal Treatment (HOT) randomised trial. HOT Study Group. Lancet 1998;351:1755-1762.

6. Benetos A, Thomas F, Bean K, Gautier S, Smulyan H, Guize L. Prognostic value of systolic and diastolic blood pressure in treated hypertensive men. Arch Intern Med 2002;162:577-581.

7. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 Report. JAMA 2003;289:2560-2572.

8. 1999 World Health Organization–International Society of Hypertension Guidelines for the Management of Hypertension. Guidelines Subcommittee. J Hypertens 1999;17:151-183.

9. Izzo JL, Jr, Levy D, Black HR. Clinical Advisory Statement. Importance of systolic blood pressure in older Americans. Hypertension 2000;35:1021-1024.

Issue
The Journal of Family Practice - 53(2)
Issue
The Journal of Family Practice - 53(2)
Page Number
138-156
Page Number
138-156
Publications
Publications
Topics
Article Type
Display Headline
Does lowering diastolic BP to less than 90 mm Hg decrease cardiovascular risk?
Display Headline
Does lowering diastolic BP to less than 90 mm Hg decrease cardiovascular risk?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

Is combining ACE inhibitors and ARBs helpful or harmful?

Article Type
Changed
Mon, 01/14/2019 - 13:11
Display Headline
Is combining ACE inhibitors and ARBs helpful or harmful?
EVIDENCE-BASED ANSWER

The combination of angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) has been studied for treatment of heart failure, hypertension, and proteinuric renal disease. Combination therapy with an ACE inhibitor and an ARB decreases symptoms in heart failure patients, but does not appear to have an impact on overall mortality (strength of recommendation [SOR]: A).

Preliminary data from small trials indicate that combination therapy may be more effective than monotherapy with an ACE inhibitor or an ARB for lowering blood pressure (SOR: B), although morbidity and mortality data for the combination are not currently available. Additionally, in trials involving diabetic and nondiabetic proteinuric renal disease, the combination of ACE inhibitors and ARBs delays progression of renal disease to a greater extent than monotherapy; however, mortality data are also unavailable (SOR: A).

 

Evidence summary

ACE inhibitors have been used most commonly for the treatment of congestive heart failure and hypertension and to slow the progression of proteinuria. Their primary mechanism of action is the suppression of angiotensin II by blocking its formation via renin and angiotensin I, thereby reducing the main deleterious effects of angiotensin II, which are mediated through vaso-constriction. Other pathways of angiotensin II formation exist and may escape inhibition of the converting enzyme.1 ARBs block the action of angiotensin II at the AT1 receptor and may, in theory, provide additive benefit.

The data describing the use of the combination of an ACE inhibitor and an ARB in heart failure are from the Valsartan Heart Failure Trial (ValHeFT),2 the Candesartan in Heart Failure Assessment of Reduction in Mortality and Morbidity Trial (CHARM),3 and in the Valsartan in Acute Myocardial Infarction Trial (VALIANT).4

In ValHeFT, 5010 patients with systolic dysfunction were randomized to the ARB valsartan or placebo in addition to background therapy, which included an ACE inhibitor in 93% of subjects. The primary endpoints were mortality and combined mortality and morbidity. An increase in mortality was found among patients on the triple therapy combination of valsartan, an ACE inhibitor, and a beta-blocker (relative risk [RR]=1.4; 95% confidence interval [CI], 1.1–1.9). Among those not on beta-blockers, adding valsartan to baseline therapy of an ACE inhibitor resulted in a modest improvement in the combined endpoint (RR=0.8; 95% CI, 0.7–0.9), but no change in mortality alone was found.2

In CHARM, candesartan was added to baseline therapy among patients with heart failure. Baseline therapy included diuretics (90%), beta blockers (55%), spironolactone (17%), and other cardiovascular medications as necessary. In this study, those in the treatment arm had a decrease in the combined endpoint of cardiovascular death plus congestive heart failure admission (RR=0.85; 95% CI, 0.75–0.96), but no difference was seen in overall mortality. Of note, no adverse interaction was demonstrated for those on the triple combination of ACE inhibitors, ARBs, and beta-blockers.3

Similarly, VALIANT demonstrated the safety but the lack of incremental efficacy in adding valsartan to ACE inhibitors for patients with left ventricular dysfunction after a myocardial infarction.4

Limited evidence is available from randomized controlled trials on the safety or efficacy of combination therapy exclusively for hyperten-sive patients. The available published trials were short-term and assessed blood pressure rather than more clinically significant endpoints such as risk of cardiovascular events and mortality. One trial of 177 patients found no significant difference in 24-hour ambulatory mean diastolic blood pressure with combination therapy vs ACE inhibitor or ARB monotherapy, but did show a decrease in clinic diastolic blood pressure.5 Another small trial of 20 patients demonstrated improved ambulatory blood pressure control with combination therapy vs ACE inhibitor monotherapy.6

 

 

 

Several trials have investigated the effect of combination therapy on diabetic and nondiabet-ic proteinuria. Conclusions from these trials are limited by their small sample size and by measurement of intermediate outcomes without mortality data. The largest trial, COOPERATE, was conducted in Japan and included 336 patients with nondiabetic renal disease.7 The investigators found that significantly fewer patients receiving combination therapy reached the combined primary endpoint of time to doubling of serum creatinine or end-stage renal disease compared with patients receiving monotherapy. The CALM study included 199 patients with hypertension, micro-albuminuria, and type 2 diabetes mellitus, and demonstrated significantly greater attenuation of urinary albumin/creatinine ratio and significantly improved blood pressure control with combination therapy compared with either therapy alone.8

Another trial, ONTARGET, is being conducted to assess the impact of ACE inhibitor or ARB monotherapy and combination therapy on reducing cardiovascular risk; it includes a combined primary endpoint of morbidity and mortality. The study involves 23,400 high-risk patients and will have a follow-up period of 5.5 years. This trial enrolls patients who have coronary disease, cere-brovascular disease, peripheral vascular disease, or diabetes with end-organ damage (inclusion and exclusion criteria are based upon those used in the HOPE study).

Recommendations from others

We were unable to find to find any recommendations regarding the addition of ARB drugs to ACE inhibitors.

CLINICAL COMMENTARY

Adding ARBs to ACE inhibitors: Good in theory, but clinical evidence is still weak
David Kilgore, MD
Tacoma Family Medicine, Tacoma, Wash

There is good evidence of the benefits of angiotensin inhibition in multiple diseases, so it is logical to ask if adding receptor blockers adds further benefit. For now, it appears that the addition of an ARB to an ACE inhibitor is an idea that sounds good in theory, but needs more data to prove its clinical benefit and safety.

The clinical evidence for the combo in heart failure and hypertension is weak, since mortality data are lacking and there is the troubling association with increased mortality in the presence of beta blockers. Using the combination is not currently recommended by the major national guidelines for those areas (eg, American Heart Association, Joint National Committee VII). Although the benefit for patients with proteinuria appears promising, we still await evidence for decreasing mortality. Given cost and the combination’s uncertain benefit, it would be prudent to wait until the completion of studies currently in progress before we embrace it.

References

1. Baruch L, Anand I, Cohen IS, Ziesche S, Judd D, Cohn JN. Augmented short- and long-term hemodynamic and hormonal effects of an angiotensin receptor blocker added to angiotensin converting enzyme inhibitor therapy in patients with heart failure. Vasodilator Heart Failure Trial (V-HeFT) Study Group. Circulation 1999;99:2658-2664.

2. Cohn JN, Tognoni G, Valsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure. N Engl J Med 2001;345:1667-1675.

3. McMurray JJ, Ostergren J, Swedberg K, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors: the CHARM-Added trial. Lancet 2003;362:767-771.

4. Pfeffer MA, McMurray JJ, Velazquez EJ, et al. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med 2003;349:1893-1906.

5. Azizi M, Linhart A, Alexander J, et al. Pilot study of combined blockade of the renin-angiotensin system in essential hypertensive patients. J Hypertens 2000;18:1139-1147.

6. Stergiou GS, Skeva II, Baibas NM, et al. Additive hypotensive effect of angiotensin-converting enzyme inhibition and angiotensin-receptor antagonism in essential hypertension. J Cardiovasc Pharmacol 2000;35:937-941.

7. Nakao N, Yoshimura A, Morita H, Takada M, Kayano T, Ideura T. Combination treatment of angiotensin-II receptor blocker and angiotensin-converting-enzyme inhibitor in non-diabetic renal disease (COOPERATE): a randomised controlled trial. Lancet 2003;361:117-124.

8. Mogensen CE, Neldam S, Tikkanen I, et al. Randomised controlled trial of dual blockade of the renin-angiotensin system in patients with hypertension, microalbuminuria, and non-insulin dependent diabetes: the candesartan and lisinopril microalbuminuria (CALM) study. BMJ 2000;321:1440-1444.

Article PDF
Author and Disclosure Information

William E. Chavey, MD, MS
Department of Family Medicine, University of Michigan, Ypsilanti;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Issue
The Journal of Family Practice - 53(2)
Publications
Topics
Page Number
138-156
Sections
Author and Disclosure Information

William E. Chavey, MD, MS
Department of Family Medicine, University of Michigan, Ypsilanti;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Author and Disclosure Information

William E. Chavey, MD, MS
Department of Family Medicine, University of Michigan, Ypsilanti;

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

The combination of angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) has been studied for treatment of heart failure, hypertension, and proteinuric renal disease. Combination therapy with an ACE inhibitor and an ARB decreases symptoms in heart failure patients, but does not appear to have an impact on overall mortality (strength of recommendation [SOR]: A).

Preliminary data from small trials indicate that combination therapy may be more effective than monotherapy with an ACE inhibitor or an ARB for lowering blood pressure (SOR: B), although morbidity and mortality data for the combination are not currently available. Additionally, in trials involving diabetic and nondiabetic proteinuric renal disease, the combination of ACE inhibitors and ARBs delays progression of renal disease to a greater extent than monotherapy; however, mortality data are also unavailable (SOR: A).

 

Evidence summary

ACE inhibitors have been used most commonly for the treatment of congestive heart failure and hypertension and to slow the progression of proteinuria. Their primary mechanism of action is the suppression of angiotensin II by blocking its formation via renin and angiotensin I, thereby reducing the main deleterious effects of angiotensin II, which are mediated through vaso-constriction. Other pathways of angiotensin II formation exist and may escape inhibition of the converting enzyme.1 ARBs block the action of angiotensin II at the AT1 receptor and may, in theory, provide additive benefit.

The data describing the use of the combination of an ACE inhibitor and an ARB in heart failure are from the Valsartan Heart Failure Trial (ValHeFT),2 the Candesartan in Heart Failure Assessment of Reduction in Mortality and Morbidity Trial (CHARM),3 and in the Valsartan in Acute Myocardial Infarction Trial (VALIANT).4

In ValHeFT, 5010 patients with systolic dysfunction were randomized to the ARB valsartan or placebo in addition to background therapy, which included an ACE inhibitor in 93% of subjects. The primary endpoints were mortality and combined mortality and morbidity. An increase in mortality was found among patients on the triple therapy combination of valsartan, an ACE inhibitor, and a beta-blocker (relative risk [RR]=1.4; 95% confidence interval [CI], 1.1–1.9). Among those not on beta-blockers, adding valsartan to baseline therapy of an ACE inhibitor resulted in a modest improvement in the combined endpoint (RR=0.8; 95% CI, 0.7–0.9), but no change in mortality alone was found.2

In CHARM, candesartan was added to baseline therapy among patients with heart failure. Baseline therapy included diuretics (90%), beta blockers (55%), spironolactone (17%), and other cardiovascular medications as necessary. In this study, those in the treatment arm had a decrease in the combined endpoint of cardiovascular death plus congestive heart failure admission (RR=0.85; 95% CI, 0.75–0.96), but no difference was seen in overall mortality. Of note, no adverse interaction was demonstrated for those on the triple combination of ACE inhibitors, ARBs, and beta-blockers.3

Similarly, VALIANT demonstrated the safety but the lack of incremental efficacy in adding valsartan to ACE inhibitors for patients with left ventricular dysfunction after a myocardial infarction.4

Limited evidence is available from randomized controlled trials on the safety or efficacy of combination therapy exclusively for hyperten-sive patients. The available published trials were short-term and assessed blood pressure rather than more clinically significant endpoints such as risk of cardiovascular events and mortality. One trial of 177 patients found no significant difference in 24-hour ambulatory mean diastolic blood pressure with combination therapy vs ACE inhibitor or ARB monotherapy, but did show a decrease in clinic diastolic blood pressure.5 Another small trial of 20 patients demonstrated improved ambulatory blood pressure control with combination therapy vs ACE inhibitor monotherapy.6

 

 

 

Several trials have investigated the effect of combination therapy on diabetic and nondiabet-ic proteinuria. Conclusions from these trials are limited by their small sample size and by measurement of intermediate outcomes without mortality data. The largest trial, COOPERATE, was conducted in Japan and included 336 patients with nondiabetic renal disease.7 The investigators found that significantly fewer patients receiving combination therapy reached the combined primary endpoint of time to doubling of serum creatinine or end-stage renal disease compared with patients receiving monotherapy. The CALM study included 199 patients with hypertension, micro-albuminuria, and type 2 diabetes mellitus, and demonstrated significantly greater attenuation of urinary albumin/creatinine ratio and significantly improved blood pressure control with combination therapy compared with either therapy alone.8

Another trial, ONTARGET, is being conducted to assess the impact of ACE inhibitor or ARB monotherapy and combination therapy on reducing cardiovascular risk; it includes a combined primary endpoint of morbidity and mortality. The study involves 23,400 high-risk patients and will have a follow-up period of 5.5 years. This trial enrolls patients who have coronary disease, cere-brovascular disease, peripheral vascular disease, or diabetes with end-organ damage (inclusion and exclusion criteria are based upon those used in the HOPE study).

Recommendations from others

We were unable to find to find any recommendations regarding the addition of ARB drugs to ACE inhibitors.

CLINICAL COMMENTARY

Adding ARBs to ACE inhibitors: Good in theory, but clinical evidence is still weak
David Kilgore, MD
Tacoma Family Medicine, Tacoma, Wash

There is good evidence of the benefits of angiotensin inhibition in multiple diseases, so it is logical to ask if adding receptor blockers adds further benefit. For now, it appears that the addition of an ARB to an ACE inhibitor is an idea that sounds good in theory, but needs more data to prove its clinical benefit and safety.

The clinical evidence for the combo in heart failure and hypertension is weak, since mortality data are lacking and there is the troubling association with increased mortality in the presence of beta blockers. Using the combination is not currently recommended by the major national guidelines for those areas (eg, American Heart Association, Joint National Committee VII). Although the benefit for patients with proteinuria appears promising, we still await evidence for decreasing mortality. Given cost and the combination’s uncertain benefit, it would be prudent to wait until the completion of studies currently in progress before we embrace it.

EVIDENCE-BASED ANSWER

The combination of angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) has been studied for treatment of heart failure, hypertension, and proteinuric renal disease. Combination therapy with an ACE inhibitor and an ARB decreases symptoms in heart failure patients, but does not appear to have an impact on overall mortality (strength of recommendation [SOR]: A).

Preliminary data from small trials indicate that combination therapy may be more effective than monotherapy with an ACE inhibitor or an ARB for lowering blood pressure (SOR: B), although morbidity and mortality data for the combination are not currently available. Additionally, in trials involving diabetic and nondiabetic proteinuric renal disease, the combination of ACE inhibitors and ARBs delays progression of renal disease to a greater extent than monotherapy; however, mortality data are also unavailable (SOR: A).

 

Evidence summary

ACE inhibitors have been used most commonly for the treatment of congestive heart failure and hypertension and to slow the progression of proteinuria. Their primary mechanism of action is the suppression of angiotensin II by blocking its formation via renin and angiotensin I, thereby reducing the main deleterious effects of angiotensin II, which are mediated through vaso-constriction. Other pathways of angiotensin II formation exist and may escape inhibition of the converting enzyme.1 ARBs block the action of angiotensin II at the AT1 receptor and may, in theory, provide additive benefit.

The data describing the use of the combination of an ACE inhibitor and an ARB in heart failure are from the Valsartan Heart Failure Trial (ValHeFT),2 the Candesartan in Heart Failure Assessment of Reduction in Mortality and Morbidity Trial (CHARM),3 and in the Valsartan in Acute Myocardial Infarction Trial (VALIANT).4

In ValHeFT, 5010 patients with systolic dysfunction were randomized to the ARB valsartan or placebo in addition to background therapy, which included an ACE inhibitor in 93% of subjects. The primary endpoints were mortality and combined mortality and morbidity. An increase in mortality was found among patients on the triple therapy combination of valsartan, an ACE inhibitor, and a beta-blocker (relative risk [RR]=1.4; 95% confidence interval [CI], 1.1–1.9). Among those not on beta-blockers, adding valsartan to baseline therapy of an ACE inhibitor resulted in a modest improvement in the combined endpoint (RR=0.8; 95% CI, 0.7–0.9), but no change in mortality alone was found.2

In CHARM, candesartan was added to baseline therapy among patients with heart failure. Baseline therapy included diuretics (90%), beta blockers (55%), spironolactone (17%), and other cardiovascular medications as necessary. In this study, those in the treatment arm had a decrease in the combined endpoint of cardiovascular death plus congestive heart failure admission (RR=0.85; 95% CI, 0.75–0.96), but no difference was seen in overall mortality. Of note, no adverse interaction was demonstrated for those on the triple combination of ACE inhibitors, ARBs, and beta-blockers.3

Similarly, VALIANT demonstrated the safety but the lack of incremental efficacy in adding valsartan to ACE inhibitors for patients with left ventricular dysfunction after a myocardial infarction.4

Limited evidence is available from randomized controlled trials on the safety or efficacy of combination therapy exclusively for hyperten-sive patients. The available published trials were short-term and assessed blood pressure rather than more clinically significant endpoints such as risk of cardiovascular events and mortality. One trial of 177 patients found no significant difference in 24-hour ambulatory mean diastolic blood pressure with combination therapy vs ACE inhibitor or ARB monotherapy, but did show a decrease in clinic diastolic blood pressure.5 Another small trial of 20 patients demonstrated improved ambulatory blood pressure control with combination therapy vs ACE inhibitor monotherapy.6

 

 

 

Several trials have investigated the effect of combination therapy on diabetic and nondiabet-ic proteinuria. Conclusions from these trials are limited by their small sample size and by measurement of intermediate outcomes without mortality data. The largest trial, COOPERATE, was conducted in Japan and included 336 patients with nondiabetic renal disease.7 The investigators found that significantly fewer patients receiving combination therapy reached the combined primary endpoint of time to doubling of serum creatinine or end-stage renal disease compared with patients receiving monotherapy. The CALM study included 199 patients with hypertension, micro-albuminuria, and type 2 diabetes mellitus, and demonstrated significantly greater attenuation of urinary albumin/creatinine ratio and significantly improved blood pressure control with combination therapy compared with either therapy alone.8

Another trial, ONTARGET, is being conducted to assess the impact of ACE inhibitor or ARB monotherapy and combination therapy on reducing cardiovascular risk; it includes a combined primary endpoint of morbidity and mortality. The study involves 23,400 high-risk patients and will have a follow-up period of 5.5 years. This trial enrolls patients who have coronary disease, cere-brovascular disease, peripheral vascular disease, or diabetes with end-organ damage (inclusion and exclusion criteria are based upon those used in the HOPE study).

Recommendations from others

We were unable to find to find any recommendations regarding the addition of ARB drugs to ACE inhibitors.

CLINICAL COMMENTARY

Adding ARBs to ACE inhibitors: Good in theory, but clinical evidence is still weak
David Kilgore, MD
Tacoma Family Medicine, Tacoma, Wash

There is good evidence of the benefits of angiotensin inhibition in multiple diseases, so it is logical to ask if adding receptor blockers adds further benefit. For now, it appears that the addition of an ARB to an ACE inhibitor is an idea that sounds good in theory, but needs more data to prove its clinical benefit and safety.

The clinical evidence for the combo in heart failure and hypertension is weak, since mortality data are lacking and there is the troubling association with increased mortality in the presence of beta blockers. Using the combination is not currently recommended by the major national guidelines for those areas (eg, American Heart Association, Joint National Committee VII). Although the benefit for patients with proteinuria appears promising, we still await evidence for decreasing mortality. Given cost and the combination’s uncertain benefit, it would be prudent to wait until the completion of studies currently in progress before we embrace it.

References

1. Baruch L, Anand I, Cohen IS, Ziesche S, Judd D, Cohn JN. Augmented short- and long-term hemodynamic and hormonal effects of an angiotensin receptor blocker added to angiotensin converting enzyme inhibitor therapy in patients with heart failure. Vasodilator Heart Failure Trial (V-HeFT) Study Group. Circulation 1999;99:2658-2664.

2. Cohn JN, Tognoni G, Valsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure. N Engl J Med 2001;345:1667-1675.

3. McMurray JJ, Ostergren J, Swedberg K, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors: the CHARM-Added trial. Lancet 2003;362:767-771.

4. Pfeffer MA, McMurray JJ, Velazquez EJ, et al. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med 2003;349:1893-1906.

5. Azizi M, Linhart A, Alexander J, et al. Pilot study of combined blockade of the renin-angiotensin system in essential hypertensive patients. J Hypertens 2000;18:1139-1147.

6. Stergiou GS, Skeva II, Baibas NM, et al. Additive hypotensive effect of angiotensin-converting enzyme inhibition and angiotensin-receptor antagonism in essential hypertension. J Cardiovasc Pharmacol 2000;35:937-941.

7. Nakao N, Yoshimura A, Morita H, Takada M, Kayano T, Ideura T. Combination treatment of angiotensin-II receptor blocker and angiotensin-converting-enzyme inhibitor in non-diabetic renal disease (COOPERATE): a randomised controlled trial. Lancet 2003;361:117-124.

8. Mogensen CE, Neldam S, Tikkanen I, et al. Randomised controlled trial of dual blockade of the renin-angiotensin system in patients with hypertension, microalbuminuria, and non-insulin dependent diabetes: the candesartan and lisinopril microalbuminuria (CALM) study. BMJ 2000;321:1440-1444.

References

1. Baruch L, Anand I, Cohen IS, Ziesche S, Judd D, Cohn JN. Augmented short- and long-term hemodynamic and hormonal effects of an angiotensin receptor blocker added to angiotensin converting enzyme inhibitor therapy in patients with heart failure. Vasodilator Heart Failure Trial (V-HeFT) Study Group. Circulation 1999;99:2658-2664.

2. Cohn JN, Tognoni G, Valsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure. N Engl J Med 2001;345:1667-1675.

3. McMurray JJ, Ostergren J, Swedberg K, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors: the CHARM-Added trial. Lancet 2003;362:767-771.

4. Pfeffer MA, McMurray JJ, Velazquez EJ, et al. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med 2003;349:1893-1906.

5. Azizi M, Linhart A, Alexander J, et al. Pilot study of combined blockade of the renin-angiotensin system in essential hypertensive patients. J Hypertens 2000;18:1139-1147.

6. Stergiou GS, Skeva II, Baibas NM, et al. Additive hypotensive effect of angiotensin-converting enzyme inhibition and angiotensin-receptor antagonism in essential hypertension. J Cardiovasc Pharmacol 2000;35:937-941.

7. Nakao N, Yoshimura A, Morita H, Takada M, Kayano T, Ideura T. Combination treatment of angiotensin-II receptor blocker and angiotensin-converting-enzyme inhibitor in non-diabetic renal disease (COOPERATE): a randomised controlled trial. Lancet 2003;361:117-124.

8. Mogensen CE, Neldam S, Tikkanen I, et al. Randomised controlled trial of dual blockade of the renin-angiotensin system in patients with hypertension, microalbuminuria, and non-insulin dependent diabetes: the candesartan and lisinopril microalbuminuria (CALM) study. BMJ 2000;321:1440-1444.

Issue
The Journal of Family Practice - 53(2)
Issue
The Journal of Family Practice - 53(2)
Page Number
138-156
Page Number
138-156
Publications
Publications
Topics
Article Type
Display Headline
Is combining ACE inhibitors and ARBs helpful or harmful?
Display Headline
Is combining ACE inhibitors and ARBs helpful or harmful?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

When should we treat isolated high triglycerides?

Article Type
Changed
Mon, 01/14/2019 - 10:59
Display Headline
When should we treat isolated high triglycerides?
EVIDENCE-BASED ANSWER

No evidence exists that treating isolated high triglyceride levels in the absence of other risk factors prevents coronary events. Although elevated triglycerides in some studies correlates with coronary events, the association weakens when controlled for factors such as diabetes, high-density lipoprotein (HDL) and low-density lipoprotein (LDL) cholesterol, body mass index, and other cardiac risk factors.

Coincident lowering of triglycerides, while treating other dyslipidemias (such as high LDL and low HDL), can contribute to decreasing coronary events (strength of recommendation [SOR]: A, based randomized controlled trials). Treating triglyceride levels over 500 to 1000 mg/dL may reduce the risk of pancreatitis (SOR: C, expert opinion).

 

Evidence summary

Truly isolated hypertriglyceridemia is rare. To date, no good trials directly address the effect of reducing truly isolated hypertriglyceridemia on cardiovascular morbidity or mortality. High triglycerides are usually accompanied by other features of the “metabolic syndrome” (low HDL, high LDL, insulin resistance, diabetes, hypertension, and obesity), making it almost impossible to look at these in isolation or attribute risk to a specific component.1

Whether high triglyceride levels pose risk in the true absence of these other metabolic factors is controversial. One meta-analysis of 17 population-based prospective studies of triglycerides and cardiovascular disease (including 57,000 patients) showed high triglyceride levels to be predictive of cardiac events, even when adjusted for HDL and other risk factors (age, total and LDL cholesterol, smoking, body mass index, and blood pressure).2 After adjusting for these other risk factors, the authors found an increased risk for all cardiac endpoints (myocardial infarction, death, etc) of 14% for men and 32% for women (Men: relative risk [RR]=1.14; 95% confidence interval [CI], 1.05–1.28; Women: RR=1.37; 95% CI, 1.13–1.66).

Another meta-analysis of 3 prospective intervention trials with 15,880 enrolled subjects found that triglyceride levels did not provide any clinically meaningful information about risk beyond that provided by other cholesterol subfractions.3

In treatment trials, the most impressive risk reductions come from the groups who fit the lipid triad of low HDL, high LDL, and high triglycerides. Low levels of HDL appear to interact with hypertriglyceridemia to increase coronary risk, and all studies showing improved outcomes have simultaneously increased HDL while lowering triglycerides.4-6 In 3 large-scale prospective, placebo-controlled trials (the Helsinki Heart Study, a primary prevention study, and the VA-HIT and Bezafibrate Infarction Prevention trials, both secondary prevention studies), lowering triglycerides and raising HDL concurrently improved outcomes.5 Successful dietary and medical interventions, especially with statins and fibrates, improved overall lipid profiles—not just triglyceride levels.

Accordingly, elevated triglycerides should prompt providers to rigorously identify these other risk factors for cardiovascular morbidity and mortality, which may not be immediately obvious. In the absence of such other factors, no evidence exists to guide therapy. Expert opinion7,8 supported by epidemiologic evidence9 suggests that patients with triglyceride levels of 500 to 1000 mg/dL may have an increased risk of pancreatitis. Accordingly, providers should consider therapy to lower triglycerides to less than 500 in these patients, regardless of accompanying risk factors.

Recommendations from others

The American College of Physicians, the European Society of Cardiology, and the US Preventive Services Task Force do not recommend screening for hypertriglyceridemia. Clinical guidelines of the National Cholesterol Education Program/Adult Treatment Panel III (NCEP/ATP III), American Heart Association/American College of Cardiology, and the American Diabetes Association all support LDL lowering as the primary target of therapy based on the patients risk profile.10 NCEP/ATP III has identified triglyceride levels of <150 as normal, 150–199 as borderline high, 200–499 as high, and 500 as very high.7

A patient with high triglycerides should prompt a search for components of the “meta-bolic syndrome” and secondary causes, including high dietary fat, high alcohol intake, drugs (steroids, beta-blockers, high-estrogen oral contraceptives), medical conditions (hypothyroidism, nephrosis, renal failure, liver disease, Cushing disease, and lupus), and rare familial dyslipidemias.7,10,11

CLINICAL COMMENTARY

Elevated triglyceride level? First look at the big picture
Donald C. Spencer, MD, MBA
University of North Carolina at Chapel Hill

Observing the pendulum swings of medical knowledge over time is one of the hallmarks of the experienced family physician. As a student, I was warned of the evils of high triglycerides, only to enter a period in the 1970s and 1980s of therapeutic nihilism when triglycerides were not thought to be an independent coronary risk factor.

As outlined here, the pendulum is moving toward a more complex consideration of the effect of triglycerides on heart disease—and what we should do about it. Our patients are better served when we focus on total coronary risk rather than a specific level of triglycerides. An elevated triglyceride level leads me first to look at the glucose. I have found several poorly controlled or even new diabetic patients this way. By then following the adage to “major on the majors and minor on the minors,” I have focused on glucose and LDL control to the benefit of my patients.

References

1. Forrester JS. Triglycerides: risk factor or fellow traveler? Curr Opin Cardiol 2001;16:261-264.

2. Austin MA, Hokanson JE, Edwards KL. Hypertriglyceridemia as a cardiovascular risk factor. Am J Cardiol 1998;81(4A):7B-12B.

3. Avins AL, Neuhaus JM. Do triglycerides provide meaningful information about heart disease risk? Arch Intern Med 2000;160:1937-1944.

4. Rubins HB, Robins SJ, Collins D, et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med 1999;341:410-418.

5. Secondary prevention by raising HDL cholesterol and reducing triglyceride in patients with coronary disease: the Bezafibrate Infarction Prevention (BIP) Study. Circulation 2000;102:21-27.

6. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study a randomised study. Lancet 2001;357:905-910.

7. Expert Panel on Detection. Evaluation and. Treatment of High Blood Cholesterol in Adults. Executive Summary of the Third Report of the National Cholesterol Education Program (NCEP). Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001;285:2486-2497.

8. Chait A, Brunzell JD. Chylomicronemia syndrome. Adv Intern Med 1992;37:249-273.

9. Athyros VG, Giouleme OI, Nikolaidis NL, et al. Long-term follow-up of patients with acute hypertriglyceridemia-induced pancreatitis. J Clin Gastroenterol 2002;34:472-475.

10. Breuer HW. Hypertriglyceridemia: a review of clinical relevance and treatment options: focus on cerivastatin. Curr Med Res Opin 2001;17:60-73.

11. Malloy MJ, Kane JP. A risk factor for atherosclerosis: triglyceride-rich lipoproteins. Adv Intern Med 2001;47:111-136.

Article PDF
Author and Disclosure Information

Mark Cucuzzella, MD
Peter C. Smith, MD
University of Colorado Health Sciences Center, Denver

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Issue
The Journal of Family Practice - 53(2)
Publications
Topics
Page Number
138-156
Sections
Author and Disclosure Information

Mark Cucuzzella, MD
Peter C. Smith, MD
University of Colorado Health Sciences Center, Denver

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Author and Disclosure Information

Mark Cucuzzella, MD
Peter C. Smith, MD
University of Colorado Health Sciences Center, Denver

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

No evidence exists that treating isolated high triglyceride levels in the absence of other risk factors prevents coronary events. Although elevated triglycerides in some studies correlates with coronary events, the association weakens when controlled for factors such as diabetes, high-density lipoprotein (HDL) and low-density lipoprotein (LDL) cholesterol, body mass index, and other cardiac risk factors.

Coincident lowering of triglycerides, while treating other dyslipidemias (such as high LDL and low HDL), can contribute to decreasing coronary events (strength of recommendation [SOR]: A, based randomized controlled trials). Treating triglyceride levels over 500 to 1000 mg/dL may reduce the risk of pancreatitis (SOR: C, expert opinion).

 

Evidence summary

Truly isolated hypertriglyceridemia is rare. To date, no good trials directly address the effect of reducing truly isolated hypertriglyceridemia on cardiovascular morbidity or mortality. High triglycerides are usually accompanied by other features of the “metabolic syndrome” (low HDL, high LDL, insulin resistance, diabetes, hypertension, and obesity), making it almost impossible to look at these in isolation or attribute risk to a specific component.1

Whether high triglyceride levels pose risk in the true absence of these other metabolic factors is controversial. One meta-analysis of 17 population-based prospective studies of triglycerides and cardiovascular disease (including 57,000 patients) showed high triglyceride levels to be predictive of cardiac events, even when adjusted for HDL and other risk factors (age, total and LDL cholesterol, smoking, body mass index, and blood pressure).2 After adjusting for these other risk factors, the authors found an increased risk for all cardiac endpoints (myocardial infarction, death, etc) of 14% for men and 32% for women (Men: relative risk [RR]=1.14; 95% confidence interval [CI], 1.05–1.28; Women: RR=1.37; 95% CI, 1.13–1.66).

Another meta-analysis of 3 prospective intervention trials with 15,880 enrolled subjects found that triglyceride levels did not provide any clinically meaningful information about risk beyond that provided by other cholesterol subfractions.3

In treatment trials, the most impressive risk reductions come from the groups who fit the lipid triad of low HDL, high LDL, and high triglycerides. Low levels of HDL appear to interact with hypertriglyceridemia to increase coronary risk, and all studies showing improved outcomes have simultaneously increased HDL while lowering triglycerides.4-6 In 3 large-scale prospective, placebo-controlled trials (the Helsinki Heart Study, a primary prevention study, and the VA-HIT and Bezafibrate Infarction Prevention trials, both secondary prevention studies), lowering triglycerides and raising HDL concurrently improved outcomes.5 Successful dietary and medical interventions, especially with statins and fibrates, improved overall lipid profiles—not just triglyceride levels.

Accordingly, elevated triglycerides should prompt providers to rigorously identify these other risk factors for cardiovascular morbidity and mortality, which may not be immediately obvious. In the absence of such other factors, no evidence exists to guide therapy. Expert opinion7,8 supported by epidemiologic evidence9 suggests that patients with triglyceride levels of 500 to 1000 mg/dL may have an increased risk of pancreatitis. Accordingly, providers should consider therapy to lower triglycerides to less than 500 in these patients, regardless of accompanying risk factors.

Recommendations from others

The American College of Physicians, the European Society of Cardiology, and the US Preventive Services Task Force do not recommend screening for hypertriglyceridemia. Clinical guidelines of the National Cholesterol Education Program/Adult Treatment Panel III (NCEP/ATP III), American Heart Association/American College of Cardiology, and the American Diabetes Association all support LDL lowering as the primary target of therapy based on the patients risk profile.10 NCEP/ATP III has identified triglyceride levels of <150 as normal, 150–199 as borderline high, 200–499 as high, and 500 as very high.7

A patient with high triglycerides should prompt a search for components of the “meta-bolic syndrome” and secondary causes, including high dietary fat, high alcohol intake, drugs (steroids, beta-blockers, high-estrogen oral contraceptives), medical conditions (hypothyroidism, nephrosis, renal failure, liver disease, Cushing disease, and lupus), and rare familial dyslipidemias.7,10,11

CLINICAL COMMENTARY

Elevated triglyceride level? First look at the big picture
Donald C. Spencer, MD, MBA
University of North Carolina at Chapel Hill

Observing the pendulum swings of medical knowledge over time is one of the hallmarks of the experienced family physician. As a student, I was warned of the evils of high triglycerides, only to enter a period in the 1970s and 1980s of therapeutic nihilism when triglycerides were not thought to be an independent coronary risk factor.

As outlined here, the pendulum is moving toward a more complex consideration of the effect of triglycerides on heart disease—and what we should do about it. Our patients are better served when we focus on total coronary risk rather than a specific level of triglycerides. An elevated triglyceride level leads me first to look at the glucose. I have found several poorly controlled or even new diabetic patients this way. By then following the adage to “major on the majors and minor on the minors,” I have focused on glucose and LDL control to the benefit of my patients.

EVIDENCE-BASED ANSWER

No evidence exists that treating isolated high triglyceride levels in the absence of other risk factors prevents coronary events. Although elevated triglycerides in some studies correlates with coronary events, the association weakens when controlled for factors such as diabetes, high-density lipoprotein (HDL) and low-density lipoprotein (LDL) cholesterol, body mass index, and other cardiac risk factors.

Coincident lowering of triglycerides, while treating other dyslipidemias (such as high LDL and low HDL), can contribute to decreasing coronary events (strength of recommendation [SOR]: A, based randomized controlled trials). Treating triglyceride levels over 500 to 1000 mg/dL may reduce the risk of pancreatitis (SOR: C, expert opinion).

 

Evidence summary

Truly isolated hypertriglyceridemia is rare. To date, no good trials directly address the effect of reducing truly isolated hypertriglyceridemia on cardiovascular morbidity or mortality. High triglycerides are usually accompanied by other features of the “metabolic syndrome” (low HDL, high LDL, insulin resistance, diabetes, hypertension, and obesity), making it almost impossible to look at these in isolation or attribute risk to a specific component.1

Whether high triglyceride levels pose risk in the true absence of these other metabolic factors is controversial. One meta-analysis of 17 population-based prospective studies of triglycerides and cardiovascular disease (including 57,000 patients) showed high triglyceride levels to be predictive of cardiac events, even when adjusted for HDL and other risk factors (age, total and LDL cholesterol, smoking, body mass index, and blood pressure).2 After adjusting for these other risk factors, the authors found an increased risk for all cardiac endpoints (myocardial infarction, death, etc) of 14% for men and 32% for women (Men: relative risk [RR]=1.14; 95% confidence interval [CI], 1.05–1.28; Women: RR=1.37; 95% CI, 1.13–1.66).

Another meta-analysis of 3 prospective intervention trials with 15,880 enrolled subjects found that triglyceride levels did not provide any clinically meaningful information about risk beyond that provided by other cholesterol subfractions.3

In treatment trials, the most impressive risk reductions come from the groups who fit the lipid triad of low HDL, high LDL, and high triglycerides. Low levels of HDL appear to interact with hypertriglyceridemia to increase coronary risk, and all studies showing improved outcomes have simultaneously increased HDL while lowering triglycerides.4-6 In 3 large-scale prospective, placebo-controlled trials (the Helsinki Heart Study, a primary prevention study, and the VA-HIT and Bezafibrate Infarction Prevention trials, both secondary prevention studies), lowering triglycerides and raising HDL concurrently improved outcomes.5 Successful dietary and medical interventions, especially with statins and fibrates, improved overall lipid profiles—not just triglyceride levels.

Accordingly, elevated triglycerides should prompt providers to rigorously identify these other risk factors for cardiovascular morbidity and mortality, which may not be immediately obvious. In the absence of such other factors, no evidence exists to guide therapy. Expert opinion7,8 supported by epidemiologic evidence9 suggests that patients with triglyceride levels of 500 to 1000 mg/dL may have an increased risk of pancreatitis. Accordingly, providers should consider therapy to lower triglycerides to less than 500 in these patients, regardless of accompanying risk factors.

Recommendations from others

The American College of Physicians, the European Society of Cardiology, and the US Preventive Services Task Force do not recommend screening for hypertriglyceridemia. Clinical guidelines of the National Cholesterol Education Program/Adult Treatment Panel III (NCEP/ATP III), American Heart Association/American College of Cardiology, and the American Diabetes Association all support LDL lowering as the primary target of therapy based on the patients risk profile.10 NCEP/ATP III has identified triglyceride levels of <150 as normal, 150–199 as borderline high, 200–499 as high, and 500 as very high.7

A patient with high triglycerides should prompt a search for components of the “meta-bolic syndrome” and secondary causes, including high dietary fat, high alcohol intake, drugs (steroids, beta-blockers, high-estrogen oral contraceptives), medical conditions (hypothyroidism, nephrosis, renal failure, liver disease, Cushing disease, and lupus), and rare familial dyslipidemias.7,10,11

CLINICAL COMMENTARY

Elevated triglyceride level? First look at the big picture
Donald C. Spencer, MD, MBA
University of North Carolina at Chapel Hill

Observing the pendulum swings of medical knowledge over time is one of the hallmarks of the experienced family physician. As a student, I was warned of the evils of high triglycerides, only to enter a period in the 1970s and 1980s of therapeutic nihilism when triglycerides were not thought to be an independent coronary risk factor.

As outlined here, the pendulum is moving toward a more complex consideration of the effect of triglycerides on heart disease—and what we should do about it. Our patients are better served when we focus on total coronary risk rather than a specific level of triglycerides. An elevated triglyceride level leads me first to look at the glucose. I have found several poorly controlled or even new diabetic patients this way. By then following the adage to “major on the majors and minor on the minors,” I have focused on glucose and LDL control to the benefit of my patients.

References

1. Forrester JS. Triglycerides: risk factor or fellow traveler? Curr Opin Cardiol 2001;16:261-264.

2. Austin MA, Hokanson JE, Edwards KL. Hypertriglyceridemia as a cardiovascular risk factor. Am J Cardiol 1998;81(4A):7B-12B.

3. Avins AL, Neuhaus JM. Do triglycerides provide meaningful information about heart disease risk? Arch Intern Med 2000;160:1937-1944.

4. Rubins HB, Robins SJ, Collins D, et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med 1999;341:410-418.

5. Secondary prevention by raising HDL cholesterol and reducing triglyceride in patients with coronary disease: the Bezafibrate Infarction Prevention (BIP) Study. Circulation 2000;102:21-27.

6. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study a randomised study. Lancet 2001;357:905-910.

7. Expert Panel on Detection. Evaluation and. Treatment of High Blood Cholesterol in Adults. Executive Summary of the Third Report of the National Cholesterol Education Program (NCEP). Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001;285:2486-2497.

8. Chait A, Brunzell JD. Chylomicronemia syndrome. Adv Intern Med 1992;37:249-273.

9. Athyros VG, Giouleme OI, Nikolaidis NL, et al. Long-term follow-up of patients with acute hypertriglyceridemia-induced pancreatitis. J Clin Gastroenterol 2002;34:472-475.

10. Breuer HW. Hypertriglyceridemia: a review of clinical relevance and treatment options: focus on cerivastatin. Curr Med Res Opin 2001;17:60-73.

11. Malloy MJ, Kane JP. A risk factor for atherosclerosis: triglyceride-rich lipoproteins. Adv Intern Med 2001;47:111-136.

References

1. Forrester JS. Triglycerides: risk factor or fellow traveler? Curr Opin Cardiol 2001;16:261-264.

2. Austin MA, Hokanson JE, Edwards KL. Hypertriglyceridemia as a cardiovascular risk factor. Am J Cardiol 1998;81(4A):7B-12B.

3. Avins AL, Neuhaus JM. Do triglycerides provide meaningful information about heart disease risk? Arch Intern Med 2000;160:1937-1944.

4. Rubins HB, Robins SJ, Collins D, et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med 1999;341:410-418.

5. Secondary prevention by raising HDL cholesterol and reducing triglyceride in patients with coronary disease: the Bezafibrate Infarction Prevention (BIP) Study. Circulation 2000;102:21-27.

6. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study a randomised study. Lancet 2001;357:905-910.

7. Expert Panel on Detection. Evaluation and. Treatment of High Blood Cholesterol in Adults. Executive Summary of the Third Report of the National Cholesterol Education Program (NCEP). Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001;285:2486-2497.

8. Chait A, Brunzell JD. Chylomicronemia syndrome. Adv Intern Med 1992;37:249-273.

9. Athyros VG, Giouleme OI, Nikolaidis NL, et al. Long-term follow-up of patients with acute hypertriglyceridemia-induced pancreatitis. J Clin Gastroenterol 2002;34:472-475.

10. Breuer HW. Hypertriglyceridemia: a review of clinical relevance and treatment options: focus on cerivastatin. Curr Med Res Opin 2001;17:60-73.

11. Malloy MJ, Kane JP. A risk factor for atherosclerosis: triglyceride-rich lipoproteins. Adv Intern Med 2001;47:111-136.

Issue
The Journal of Family Practice - 53(2)
Issue
The Journal of Family Practice - 53(2)
Page Number
138-156
Page Number
138-156
Publications
Publications
Topics
Article Type
Display Headline
When should we treat isolated high triglycerides?
Display Headline
When should we treat isolated high triglycerides?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

How accurate is stress radionuclide imaging for diagnosis of CAD?

Article Type
Changed
Mon, 01/14/2019 - 10:59
Display Headline
How accurate is stress radionuclide imaging for diagnosis of CAD?
EVIDENCE-BASED ANSWER

Stress radionuclide testing is a moderately accurate test compared with coronary angiography for the diagnosis of coronary artery disease (CAD) in intermediate-risk individuals.

Variations in technique of imaging (planar or single-photon emission computed tomography [SPECT]) and stress (exercise or pharmacologic) do not significantly alter the accuracy of this test, although there is some evidence for decreased accuracy in women (strength of recommendation [SOR]: A, based on multiple meta-analyses). Abnormal stress radionuclide screening in vascular surgical candidates also predicts an increased rate of perioperative cardiac events (SOR: A, based on meta-analysis).

 

Evidence summary

Stress radionuclide imaging—specifically its diagnostic accuracy—has been the subject of numerous studies. Detrano et al1 reported the first pooled data (56 studies); they concluded that estimates of sensitivity (85%) and specificity (85%) are biased by studies that were not blinded, included subjects with prior myocardial infarction (MI), or had a work-up (verification) bias (ie, use of the gold standard test is affected by the result on the test under question).

Another systematic review reported estimates of sensitivity ranging from 68% to 96% and specificity from 65% to 100%.2 The review was accompanied by a position paper from the American College of Physicians stating that the test may be appropriate for a patient with intermediate risk of coronary artery disease.3

Four meta-analyses report diagnostic accuracy of radionuclide cardiac imaging (Table). Kwok et al6 analyzed data on women only and found decreased diagnostic accuracy in this population. Kim et al7 analyzed pharmacologic stressors used with SPECT and confirmed that accuracy is near that of exercise SPECT. Patient-centered outcomes were reported in a meta-analysis of dipyridamole-thallium imaging in the preoperative evaluation of vascular surgery patients. The summary odds ratio for any perioperative cardiac event (in patients with abnormal tests) was 3.5 (95% confidence interval [CI], 2.5–4.8); the odds ratio for MI or cardiac death was 3.9 (95% CI, 2.5–5.6), leading the authors to conclude that there is sound evidence to use radionuclide testing in intermedi.ate-risk patients during preoperative screening.8

TABLE
Diagnostic accuracy reported in meta-analyses of cardiac radionuclide SPECT imaging

Authors, yearStudiesSn % (95% CI)Sp % (95% CI)LR+LR–
Garber and Solomon 19944888 (73–98)77 (53–96)3.80.16
Fleischmann et al, et al 199852787 (86–88)64 (60–68)2.40.20
Kwok et al, 19966378 (69–87)58 (51–66)1.90.38
Kim et al, 200174490 (89–92)*75 (70–79)*3.60.13
89 (84–93)65 (54–74)2.50.17
82(77–87)73 (70–79)3.00.25
*Adenosine SPECT
†Dipyridamole SPECT
‡Dobutamine SPECT
SPECT, single-photon emission computed tomography; SN, sensitivity; Sp, specifity; LR+, positive likelihood ratio; LR–, negative likelihood ratio; Cl, confidence interval

Recommendations from others

The American Heart Association/American College Cardiology (AHA/ACC) Task Force on Assessment of Diagnostic and Therapeutic Cardiovascular Procedures and the American Society of Nuclear Cardiology updated guidelines for cardiac radionuclide imaging in 2003. In this consensus statement (a nonsystematic review of literature and expert opinion), they reported test characteristics to detect a 50% angiographic lesion as follows—exercise SPECT: sensitivity 87%, specificity 73%; vasodilator (adenosine or dipyridamole) SPECT: sensitivity 89%, specificity 75%. They noted that quantitative analysis performs as well as qualitative analysis of radionuclide images. Gated SPECT is slightly more specific and just as sensitive as nongated SPECT.

The Taskforce recommended that radionuclide perfusion scans be performed in patients with baseline electrocardiogram (ECG) abnormalities (such as left bundle branch block, hypertrophy, digitalis effect, etc), patients who cannot perform an exercise stress test, and to assess the functional effect of indeterminate lesions found on angiography. They also note that the repeat use of radionuclide testing 3 to 5 years after an event in asymptomatic high-risk patients and the initial use of radionuclide testing in patients at very high risk are both somewhat controversial, but the weight of limited evidence suggests some benefit to their use.9

CLINICAL COMMENTARY

ECG stress still the choice; image those with abnormal ECG or unable to exercise
David Kilgore, MD
Tacoma Family Medicine, Tacoma, Wash

Primary care providers frequently face the question of how best to evaluate patients with suspected CAD. Recent studies and expert opinion appear to give conflicting advice regarding the merits of plain exercise ECG vs stress imaging. Information on accuracy doesn’t always indicate which test is best for a patient.

Though quoted sensitivities and specificities for exercise ECG typically appear lower than those for stress imaging, costs for stress imaging are significantly higher, and numerous recent studies are demonstrating mortality outcome differences obtainable from physiologic information found in exercise testing (exercise capacity, blood pressure and pulse changes, time to angina).

Currently, the best choice for evaluation appears to be summarized by the 2003 AHA/ACC practice guidelines, which endorse exercise ECG for patients (women included) with intermediate pretest risk, and normal resting ECG for those who are unable to exercise. Stress imaging is cost effective for those patients with abnormal baseline ECG (left bundle branch block, ST abnormalities), or who are unable to exercise.

References

1. Detrano R, Janosi A, Lyons KP, Marcondes G, Abbassi N, Froelicher VF. Factors affecting sensitivity and specificity of a diagnostic test: the exercise thallium scintigram. Am J Med 1988;84:699-710.

2. Kotler TS, Diamond GA. Exercise thallium-201 scintigraphy in the diagnosis and prognosis of coronary artery disease. Ann Intern Med 1990;113:684-702.

3. Efficacy of exercise thallium-201 scintigraphy in diagnosis and prognosis of coronary artery disease. American College of Physicians. Ann Intern Med 1990;113:703-704.

4. Garber AM, Solomon NA. Cost-effectiveness of alternative test strategies for the diagnosis of coronary artery disease. Ann Intern Med 1999;130:719-728.

5. Fleischmann KE, Hunink MG, Kuntz KM, Douglas PS. Exercise echocardiography or exercise SPECT imaging? A meta-analysis of diagnostic test performance. JAMA 1998;280:913-920.

6. Kwok Y, Kim C, Grady D, Segal M, Redberg R. Meta-analysis of exercise testing to detect coronary artery disease in women. Am J Cardiol 1999;83:660-666.

7. Kim C, Kwok YS, Heagerty P, Redberg R. Pharmacologic stress testing for coronary artery disease diagnosis: A meta-analysis. Am Heart J 2001;142:934-944.

8. Shaw LJ, Eagle KA, Gersh BJ, Miller DD. Meta-analysis of intravenous dipyridamole-thallium-201 imaging (1985–1994) and dobutamine echocardiography (1991–1994) for risk stratification before vascular surgery. J Am Coll Cardiol 1996;27:787-798.

9. Klocke FJ, Baird MG, Bateman TM, et al. ACC/AHA/ASNC guidelines for the clinical use of cardiac radionuclide imaging: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. ACC/AHA/ASNC Committee to Revise the 1995 Guidelines for the Clinical Use of Radionuclide Imaging. Available at: www.acc.org/clinical/guidelines/radio/rni_fulltext.pdf. Accessed on December 14, 2003.

Article PDF
Author and Disclosure Information

Lynda Montgomery, MD
Department of Family Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Issue
The Journal of Family Practice - 53(1)
Publications
Topics
Page Number
55-71
Sections
Author and Disclosure Information

Lynda Montgomery, MD
Department of Family Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Author and Disclosure Information

Lynda Montgomery, MD
Department of Family Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Stress radionuclide testing is a moderately accurate test compared with coronary angiography for the diagnosis of coronary artery disease (CAD) in intermediate-risk individuals.

Variations in technique of imaging (planar or single-photon emission computed tomography [SPECT]) and stress (exercise or pharmacologic) do not significantly alter the accuracy of this test, although there is some evidence for decreased accuracy in women (strength of recommendation [SOR]: A, based on multiple meta-analyses). Abnormal stress radionuclide screening in vascular surgical candidates also predicts an increased rate of perioperative cardiac events (SOR: A, based on meta-analysis).

 

Evidence summary

Stress radionuclide imaging—specifically its diagnostic accuracy—has been the subject of numerous studies. Detrano et al1 reported the first pooled data (56 studies); they concluded that estimates of sensitivity (85%) and specificity (85%) are biased by studies that were not blinded, included subjects with prior myocardial infarction (MI), or had a work-up (verification) bias (ie, use of the gold standard test is affected by the result on the test under question).

Another systematic review reported estimates of sensitivity ranging from 68% to 96% and specificity from 65% to 100%.2 The review was accompanied by a position paper from the American College of Physicians stating that the test may be appropriate for a patient with intermediate risk of coronary artery disease.3

Four meta-analyses report diagnostic accuracy of radionuclide cardiac imaging (Table). Kwok et al6 analyzed data on women only and found decreased diagnostic accuracy in this population. Kim et al7 analyzed pharmacologic stressors used with SPECT and confirmed that accuracy is near that of exercise SPECT. Patient-centered outcomes were reported in a meta-analysis of dipyridamole-thallium imaging in the preoperative evaluation of vascular surgery patients. The summary odds ratio for any perioperative cardiac event (in patients with abnormal tests) was 3.5 (95% confidence interval [CI], 2.5–4.8); the odds ratio for MI or cardiac death was 3.9 (95% CI, 2.5–5.6), leading the authors to conclude that there is sound evidence to use radionuclide testing in intermedi.ate-risk patients during preoperative screening.8

TABLE
Diagnostic accuracy reported in meta-analyses of cardiac radionuclide SPECT imaging

Authors, yearStudiesSn % (95% CI)Sp % (95% CI)LR+LR–
Garber and Solomon 19944888 (73–98)77 (53–96)3.80.16
Fleischmann et al, et al 199852787 (86–88)64 (60–68)2.40.20
Kwok et al, 19966378 (69–87)58 (51–66)1.90.38
Kim et al, 200174490 (89–92)*75 (70–79)*3.60.13
89 (84–93)65 (54–74)2.50.17
82(77–87)73 (70–79)3.00.25
*Adenosine SPECT
†Dipyridamole SPECT
‡Dobutamine SPECT
SPECT, single-photon emission computed tomography; SN, sensitivity; Sp, specifity; LR+, positive likelihood ratio; LR–, negative likelihood ratio; Cl, confidence interval

Recommendations from others

The American Heart Association/American College Cardiology (AHA/ACC) Task Force on Assessment of Diagnostic and Therapeutic Cardiovascular Procedures and the American Society of Nuclear Cardiology updated guidelines for cardiac radionuclide imaging in 2003. In this consensus statement (a nonsystematic review of literature and expert opinion), they reported test characteristics to detect a 50% angiographic lesion as follows—exercise SPECT: sensitivity 87%, specificity 73%; vasodilator (adenosine or dipyridamole) SPECT: sensitivity 89%, specificity 75%. They noted that quantitative analysis performs as well as qualitative analysis of radionuclide images. Gated SPECT is slightly more specific and just as sensitive as nongated SPECT.

The Taskforce recommended that radionuclide perfusion scans be performed in patients with baseline electrocardiogram (ECG) abnormalities (such as left bundle branch block, hypertrophy, digitalis effect, etc), patients who cannot perform an exercise stress test, and to assess the functional effect of indeterminate lesions found on angiography. They also note that the repeat use of radionuclide testing 3 to 5 years after an event in asymptomatic high-risk patients and the initial use of radionuclide testing in patients at very high risk are both somewhat controversial, but the weight of limited evidence suggests some benefit to their use.9

CLINICAL COMMENTARY

ECG stress still the choice; image those with abnormal ECG or unable to exercise
David Kilgore, MD
Tacoma Family Medicine, Tacoma, Wash

Primary care providers frequently face the question of how best to evaluate patients with suspected CAD. Recent studies and expert opinion appear to give conflicting advice regarding the merits of plain exercise ECG vs stress imaging. Information on accuracy doesn’t always indicate which test is best for a patient.

Though quoted sensitivities and specificities for exercise ECG typically appear lower than those for stress imaging, costs for stress imaging are significantly higher, and numerous recent studies are demonstrating mortality outcome differences obtainable from physiologic information found in exercise testing (exercise capacity, blood pressure and pulse changes, time to angina).

Currently, the best choice for evaluation appears to be summarized by the 2003 AHA/ACC practice guidelines, which endorse exercise ECG for patients (women included) with intermediate pretest risk, and normal resting ECG for those who are unable to exercise. Stress imaging is cost effective for those patients with abnormal baseline ECG (left bundle branch block, ST abnormalities), or who are unable to exercise.

EVIDENCE-BASED ANSWER

Stress radionuclide testing is a moderately accurate test compared with coronary angiography for the diagnosis of coronary artery disease (CAD) in intermediate-risk individuals.

Variations in technique of imaging (planar or single-photon emission computed tomography [SPECT]) and stress (exercise or pharmacologic) do not significantly alter the accuracy of this test, although there is some evidence for decreased accuracy in women (strength of recommendation [SOR]: A, based on multiple meta-analyses). Abnormal stress radionuclide screening in vascular surgical candidates also predicts an increased rate of perioperative cardiac events (SOR: A, based on meta-analysis).

 

Evidence summary

Stress radionuclide imaging—specifically its diagnostic accuracy—has been the subject of numerous studies. Detrano et al1 reported the first pooled data (56 studies); they concluded that estimates of sensitivity (85%) and specificity (85%) are biased by studies that were not blinded, included subjects with prior myocardial infarction (MI), or had a work-up (verification) bias (ie, use of the gold standard test is affected by the result on the test under question).

Another systematic review reported estimates of sensitivity ranging from 68% to 96% and specificity from 65% to 100%.2 The review was accompanied by a position paper from the American College of Physicians stating that the test may be appropriate for a patient with intermediate risk of coronary artery disease.3

Four meta-analyses report diagnostic accuracy of radionuclide cardiac imaging (Table). Kwok et al6 analyzed data on women only and found decreased diagnostic accuracy in this population. Kim et al7 analyzed pharmacologic stressors used with SPECT and confirmed that accuracy is near that of exercise SPECT. Patient-centered outcomes were reported in a meta-analysis of dipyridamole-thallium imaging in the preoperative evaluation of vascular surgery patients. The summary odds ratio for any perioperative cardiac event (in patients with abnormal tests) was 3.5 (95% confidence interval [CI], 2.5–4.8); the odds ratio for MI or cardiac death was 3.9 (95% CI, 2.5–5.6), leading the authors to conclude that there is sound evidence to use radionuclide testing in intermedi.ate-risk patients during preoperative screening.8

TABLE
Diagnostic accuracy reported in meta-analyses of cardiac radionuclide SPECT imaging

Authors, yearStudiesSn % (95% CI)Sp % (95% CI)LR+LR–
Garber and Solomon 19944888 (73–98)77 (53–96)3.80.16
Fleischmann et al, et al 199852787 (86–88)64 (60–68)2.40.20
Kwok et al, 19966378 (69–87)58 (51–66)1.90.38
Kim et al, 200174490 (89–92)*75 (70–79)*3.60.13
89 (84–93)65 (54–74)2.50.17
82(77–87)73 (70–79)3.00.25
*Adenosine SPECT
†Dipyridamole SPECT
‡Dobutamine SPECT
SPECT, single-photon emission computed tomography; SN, sensitivity; Sp, specifity; LR+, positive likelihood ratio; LR–, negative likelihood ratio; Cl, confidence interval

Recommendations from others

The American Heart Association/American College Cardiology (AHA/ACC) Task Force on Assessment of Diagnostic and Therapeutic Cardiovascular Procedures and the American Society of Nuclear Cardiology updated guidelines for cardiac radionuclide imaging in 2003. In this consensus statement (a nonsystematic review of literature and expert opinion), they reported test characteristics to detect a 50% angiographic lesion as follows—exercise SPECT: sensitivity 87%, specificity 73%; vasodilator (adenosine or dipyridamole) SPECT: sensitivity 89%, specificity 75%. They noted that quantitative analysis performs as well as qualitative analysis of radionuclide images. Gated SPECT is slightly more specific and just as sensitive as nongated SPECT.

The Taskforce recommended that radionuclide perfusion scans be performed in patients with baseline electrocardiogram (ECG) abnormalities (such as left bundle branch block, hypertrophy, digitalis effect, etc), patients who cannot perform an exercise stress test, and to assess the functional effect of indeterminate lesions found on angiography. They also note that the repeat use of radionuclide testing 3 to 5 years after an event in asymptomatic high-risk patients and the initial use of radionuclide testing in patients at very high risk are both somewhat controversial, but the weight of limited evidence suggests some benefit to their use.9

CLINICAL COMMENTARY

ECG stress still the choice; image those with abnormal ECG or unable to exercise
David Kilgore, MD
Tacoma Family Medicine, Tacoma, Wash

Primary care providers frequently face the question of how best to evaluate patients with suspected CAD. Recent studies and expert opinion appear to give conflicting advice regarding the merits of plain exercise ECG vs stress imaging. Information on accuracy doesn’t always indicate which test is best for a patient.

Though quoted sensitivities and specificities for exercise ECG typically appear lower than those for stress imaging, costs for stress imaging are significantly higher, and numerous recent studies are demonstrating mortality outcome differences obtainable from physiologic information found in exercise testing (exercise capacity, blood pressure and pulse changes, time to angina).

Currently, the best choice for evaluation appears to be summarized by the 2003 AHA/ACC practice guidelines, which endorse exercise ECG for patients (women included) with intermediate pretest risk, and normal resting ECG for those who are unable to exercise. Stress imaging is cost effective for those patients with abnormal baseline ECG (left bundle branch block, ST abnormalities), or who are unable to exercise.

References

1. Detrano R, Janosi A, Lyons KP, Marcondes G, Abbassi N, Froelicher VF. Factors affecting sensitivity and specificity of a diagnostic test: the exercise thallium scintigram. Am J Med 1988;84:699-710.

2. Kotler TS, Diamond GA. Exercise thallium-201 scintigraphy in the diagnosis and prognosis of coronary artery disease. Ann Intern Med 1990;113:684-702.

3. Efficacy of exercise thallium-201 scintigraphy in diagnosis and prognosis of coronary artery disease. American College of Physicians. Ann Intern Med 1990;113:703-704.

4. Garber AM, Solomon NA. Cost-effectiveness of alternative test strategies for the diagnosis of coronary artery disease. Ann Intern Med 1999;130:719-728.

5. Fleischmann KE, Hunink MG, Kuntz KM, Douglas PS. Exercise echocardiography or exercise SPECT imaging? A meta-analysis of diagnostic test performance. JAMA 1998;280:913-920.

6. Kwok Y, Kim C, Grady D, Segal M, Redberg R. Meta-analysis of exercise testing to detect coronary artery disease in women. Am J Cardiol 1999;83:660-666.

7. Kim C, Kwok YS, Heagerty P, Redberg R. Pharmacologic stress testing for coronary artery disease diagnosis: A meta-analysis. Am Heart J 2001;142:934-944.

8. Shaw LJ, Eagle KA, Gersh BJ, Miller DD. Meta-analysis of intravenous dipyridamole-thallium-201 imaging (1985–1994) and dobutamine echocardiography (1991–1994) for risk stratification before vascular surgery. J Am Coll Cardiol 1996;27:787-798.

9. Klocke FJ, Baird MG, Bateman TM, et al. ACC/AHA/ASNC guidelines for the clinical use of cardiac radionuclide imaging: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. ACC/AHA/ASNC Committee to Revise the 1995 Guidelines for the Clinical Use of Radionuclide Imaging. Available at: www.acc.org/clinical/guidelines/radio/rni_fulltext.pdf. Accessed on December 14, 2003.

References

1. Detrano R, Janosi A, Lyons KP, Marcondes G, Abbassi N, Froelicher VF. Factors affecting sensitivity and specificity of a diagnostic test: the exercise thallium scintigram. Am J Med 1988;84:699-710.

2. Kotler TS, Diamond GA. Exercise thallium-201 scintigraphy in the diagnosis and prognosis of coronary artery disease. Ann Intern Med 1990;113:684-702.

3. Efficacy of exercise thallium-201 scintigraphy in diagnosis and prognosis of coronary artery disease. American College of Physicians. Ann Intern Med 1990;113:703-704.

4. Garber AM, Solomon NA. Cost-effectiveness of alternative test strategies for the diagnosis of coronary artery disease. Ann Intern Med 1999;130:719-728.

5. Fleischmann KE, Hunink MG, Kuntz KM, Douglas PS. Exercise echocardiography or exercise SPECT imaging? A meta-analysis of diagnostic test performance. JAMA 1998;280:913-920.

6. Kwok Y, Kim C, Grady D, Segal M, Redberg R. Meta-analysis of exercise testing to detect coronary artery disease in women. Am J Cardiol 1999;83:660-666.

7. Kim C, Kwok YS, Heagerty P, Redberg R. Pharmacologic stress testing for coronary artery disease diagnosis: A meta-analysis. Am Heart J 2001;142:934-944.

8. Shaw LJ, Eagle KA, Gersh BJ, Miller DD. Meta-analysis of intravenous dipyridamole-thallium-201 imaging (1985–1994) and dobutamine echocardiography (1991–1994) for risk stratification before vascular surgery. J Am Coll Cardiol 1996;27:787-798.

9. Klocke FJ, Baird MG, Bateman TM, et al. ACC/AHA/ASNC guidelines for the clinical use of cardiac radionuclide imaging: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. ACC/AHA/ASNC Committee to Revise the 1995 Guidelines for the Clinical Use of Radionuclide Imaging. Available at: www.acc.org/clinical/guidelines/radio/rni_fulltext.pdf. Accessed on December 14, 2003.

Issue
The Journal of Family Practice - 53(1)
Issue
The Journal of Family Practice - 53(1)
Page Number
55-71
Page Number
55-71
Publications
Publications
Topics
Article Type
Display Headline
How accurate is stress radionuclide imaging for diagnosis of CAD?
Display Headline
How accurate is stress radionuclide imaging for diagnosis of CAD?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

Do antiarrhythmics prevent sudden death in patients with heart failure?

Article Type
Changed
Mon, 01/14/2019 - 13:09
Display Headline
Do antiarrhythmics prevent sudden death in patients with heart failure?
EVIDENCE-BASED ANSWER

Beta-blockers (class II antiarrhythmics) reduce sudden death and total mortality in patients with heart failure (strength of recommendation [SOR]: A, based on systematic reviews of randomized controlled trials). Amiodarone (class III) may reduce sudden death in heart failure (SOR: B, extrapolation from randomized controlled trials), but evidence is weak that it reduces total mortality, and it has significant side effects. Class I and other class III antiarrhythmic agents appear cause an increase in mortality due to sudden death in heart failure (SOR: B, extrapolations from randomized controlled trials).

 

Evidence summary

Antiarrhythmic agents have been studied in patients with heart failure because these persons have a high incidence of sudden death, presumably from ventricular arrhythmias. Although the implantable defibrillator is an alternative antiarrhythmic device that may be preferred for some patients, we restricted our review to pharmacologic antiarrhythmics.

The beta-blockers bisoprolol, carvedilol, and metoprolol1-3 were studied in large randomized controlled trials. The relative risk reduction (RRR) for sudden death ranged from 10% to 52% in the larger trials and 30% to 39% in meta-analyses.1-4 The absolute risk reduction (ARR) was about 2% to 3% per year for sudden death and 3% to 5% for total mortality (number needed to treat=20–33 per year).

These beta-blockers were well-tolerated, even in class IV New York Heart Association patients, and improved other endpoints. Although we cannot say whether the benefits are a class effect, they were seen with both beta-1 selective and nonselective agents.

Amiodarone was studied in 2 large randomized controlled trials enrolling patients with heart failure, in trials that included patients with or without heart failure at high risk for sudden death (usually post-myocardial infarction or with complex ventricular arrhythmias), and in meta-analyses.5-8 The largest randomized controlled trial in heart failure showed a significant ARR of 2.9% for sudden death,5 but was unblinded. The largest placebo-controlled trial in heart failure failed to detect a significant decrease in sudden death.6

Meta-analyses, weakened by heterogeneity and the inclusion of patients without heart failure, detected a significant 21% to 25% RRR for sudden death,7,8 and an ARR of 2% to 3% per year. The pooled data from the placebo-controlled heart failure trials showed nonsignificant trends: 1.6% per year ARR for sudden death, 0.6% per year for total mortality.

These possible benefits must be balanced against the risk of harm from amiodarone, including excess rates of pulmonary infiltrate (1.1% per year), thyroid dysfunction (6.8% per year), liver enzyme abnormalities (0.6% per year), neuropathy (0.3% per year), and bradycardia (1.6% per year), as well as a discontinuation rate of 41% compared with 27% for placebo.7 No evidence suggested that use of amiodarone in patients with heart failure increased mortality.

Class I antiarrhythmics and other class III agents have not been studied in heart failure trials, but were associated with increased mortality in studies of patients at high risk for ventricular arrhythmia,9,10 including patients with left ventricular dysfunction. Because this increase in mortality is thought to be due to proarrhythmic properties of the drugs, further trials in heart failure patients are unlikely to occur.

Recommendations from others

American College of Cardiology/American Heart Association (ACC/AHA),11 European Society of Cardiology (ESC),12 and Heart Failure Society of America (HFSA) guidelines13 address heart failure. ACC/AHA and ESC reports specifically mention that beta-blockers reduce sudden death. Both strongly support the use of beta-blockers in patients with heart failure.

ACC/AHA finds “conflicting evidence and/or a divergence of opinion about the usefulness/ efficacy” of amiodarone to prevent sudden death and advises: “routine use of amiodarone to prevent sudden death is not recommended.” The ESC and HFSA also recommend against routine use of amiodarone.

All 3 guidelines, however, state that for the control of symptomatic arrhythmias in heart failure, amiodarone is the antiarrhythmic agent of choice. All 3 also recommend not using class I or other class III agents in heart failure.

CLINICAL COMMENTARY

Beta-blockers reduce mortality in patients with heart failure
Joseph Saseen, PharmD, BCPS
University of Colorado Health Sciences Center, Denver

Numerous well-controlled clinical trials have conclusively demonstrated that beta-blockers reduce morbidity and mortality (including sudden death) in patients with systolic heart failure. They are considered disease-modifying agents and their use is strongly encouraged. Beta-blocker therapy must be initiated using low doses and only when patients are hemo-dynamically stable, with gradual dose titrations to prevent acute decompensation.

Evidence for amiodarone shows some reduction in sudden death, but these data are less compelling. Moreover, adverse effects and drug interactions complicate long-term amiodarone use. Use of class I (eg, flecainide, procainamide, propafenone) and other class III (sotalol) anti-arrhythmics to reduce sudden death is discouraged.

References

1. Effect of metoprolol CR/XL in chronic heart failure. Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet 1999;353:2001-2007.

2. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial Lancet 1999;353:9-13.

3. Packer M, Bristow MR, Cohn JN, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group. N Engl J Med 1996;334:1349-1355.

4. Lee S, Spencer A. Beta-blockers to reduce mortality in patients with systolic dysfunction: a meta-analysis. J Fam Pract 2001;50:499-504.

5. Doval HC, Nul DR, Grancelli HO, Perrone SV, Bortman GR, Curiel R. Randomised trial of low-dose amiodarone in severe congestive heart failure. Grupo de Estudio de la Sobrevida en la Insuficiencia Cardiaca en Argentina (GESICA). Lancet. 1994;344:493-498.

6. Singh SN, Fletcher RD, Fisher SG, et al. Amiodarone in patients with congestive heart failure and asymptomatic ventricular arrhythmia. Survival Trial of Antiarrhythmic Therapy in Congestive Heart Failure. N Engl J Med 1995;333:77-82.

7. Effect of prophylactic amiodarone on mortality after acute myocardial infarction and in congestive heart failure: meta-analysis of individual data from 6500 patients in randomised trials. Amiodarone Trials Meta-Analysis Investigators. Lancet 1997;350:1417-1424.

8. Piepoli M, Villani GO, Ponikowski P, Wright A, Flather MD, Coats AJ. Overview and meta-analysis of randomised trials of amiodarone in chronic heart failure. Int J Cardiol 1998;66:1-10.

9. Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. N Engl J Med 1991;324:781-788.

10. Connolly SJ. Meta-analysis of anti-arrhythmic drug trials. Am J Cardiol 1999;84:90R-93R.

11. Hunt SA, Baker DW, Chin MH, et al. ACC/AHA guidelines for the evaluation and management of chronic heart failure in the adult. Bethesda, Md: American College of Cardiology; 2001.

12. Priori SG, Aliot E, Blomstrom-Lundqvist C, et al. Task Force on Sudden Cardiac Death of the European Society of Cardiology. Eur Heart J 2001;22:1374-1450.

13. HFSA guidelines for management of patients with heart failure caused by left ventricular systolic dysfunction— pharmacological approaches. Heart Failure Society of merica. Pharmacotherapy 2000;20:495-522.

Article PDF
Author and Disclosure Information

Thomas A. Ball, MD
William J. Kerns, MD
Shenandoah Valley Family Practice Residency Program, Dept of Family Practice, Virginia Commonwealth University, Front Royal, Va;

Joan Nashelsky, MLS
Iowa City, IA

Issue
The Journal of Family Practice - 52(9)
Publications
Topics
Page Number
714-724
Sections
Author and Disclosure Information

Thomas A. Ball, MD
William J. Kerns, MD
Shenandoah Valley Family Practice Residency Program, Dept of Family Practice, Virginia Commonwealth University, Front Royal, Va;

Joan Nashelsky, MLS
Iowa City, IA

Author and Disclosure Information

Thomas A. Ball, MD
William J. Kerns, MD
Shenandoah Valley Family Practice Residency Program, Dept of Family Practice, Virginia Commonwealth University, Front Royal, Va;

Joan Nashelsky, MLS
Iowa City, IA

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Beta-blockers (class II antiarrhythmics) reduce sudden death and total mortality in patients with heart failure (strength of recommendation [SOR]: A, based on systematic reviews of randomized controlled trials). Amiodarone (class III) may reduce sudden death in heart failure (SOR: B, extrapolation from randomized controlled trials), but evidence is weak that it reduces total mortality, and it has significant side effects. Class I and other class III antiarrhythmic agents appear cause an increase in mortality due to sudden death in heart failure (SOR: B, extrapolations from randomized controlled trials).

 

Evidence summary

Antiarrhythmic agents have been studied in patients with heart failure because these persons have a high incidence of sudden death, presumably from ventricular arrhythmias. Although the implantable defibrillator is an alternative antiarrhythmic device that may be preferred for some patients, we restricted our review to pharmacologic antiarrhythmics.

The beta-blockers bisoprolol, carvedilol, and metoprolol1-3 were studied in large randomized controlled trials. The relative risk reduction (RRR) for sudden death ranged from 10% to 52% in the larger trials and 30% to 39% in meta-analyses.1-4 The absolute risk reduction (ARR) was about 2% to 3% per year for sudden death and 3% to 5% for total mortality (number needed to treat=20–33 per year).

These beta-blockers were well-tolerated, even in class IV New York Heart Association patients, and improved other endpoints. Although we cannot say whether the benefits are a class effect, they were seen with both beta-1 selective and nonselective agents.

Amiodarone was studied in 2 large randomized controlled trials enrolling patients with heart failure, in trials that included patients with or without heart failure at high risk for sudden death (usually post-myocardial infarction or with complex ventricular arrhythmias), and in meta-analyses.5-8 The largest randomized controlled trial in heart failure showed a significant ARR of 2.9% for sudden death,5 but was unblinded. The largest placebo-controlled trial in heart failure failed to detect a significant decrease in sudden death.6

Meta-analyses, weakened by heterogeneity and the inclusion of patients without heart failure, detected a significant 21% to 25% RRR for sudden death,7,8 and an ARR of 2% to 3% per year. The pooled data from the placebo-controlled heart failure trials showed nonsignificant trends: 1.6% per year ARR for sudden death, 0.6% per year for total mortality.

These possible benefits must be balanced against the risk of harm from amiodarone, including excess rates of pulmonary infiltrate (1.1% per year), thyroid dysfunction (6.8% per year), liver enzyme abnormalities (0.6% per year), neuropathy (0.3% per year), and bradycardia (1.6% per year), as well as a discontinuation rate of 41% compared with 27% for placebo.7 No evidence suggested that use of amiodarone in patients with heart failure increased mortality.

Class I antiarrhythmics and other class III agents have not been studied in heart failure trials, but were associated with increased mortality in studies of patients at high risk for ventricular arrhythmia,9,10 including patients with left ventricular dysfunction. Because this increase in mortality is thought to be due to proarrhythmic properties of the drugs, further trials in heart failure patients are unlikely to occur.

Recommendations from others

American College of Cardiology/American Heart Association (ACC/AHA),11 European Society of Cardiology (ESC),12 and Heart Failure Society of America (HFSA) guidelines13 address heart failure. ACC/AHA and ESC reports specifically mention that beta-blockers reduce sudden death. Both strongly support the use of beta-blockers in patients with heart failure.

ACC/AHA finds “conflicting evidence and/or a divergence of opinion about the usefulness/ efficacy” of amiodarone to prevent sudden death and advises: “routine use of amiodarone to prevent sudden death is not recommended.” The ESC and HFSA also recommend against routine use of amiodarone.

All 3 guidelines, however, state that for the control of symptomatic arrhythmias in heart failure, amiodarone is the antiarrhythmic agent of choice. All 3 also recommend not using class I or other class III agents in heart failure.

CLINICAL COMMENTARY

Beta-blockers reduce mortality in patients with heart failure
Joseph Saseen, PharmD, BCPS
University of Colorado Health Sciences Center, Denver

Numerous well-controlled clinical trials have conclusively demonstrated that beta-blockers reduce morbidity and mortality (including sudden death) in patients with systolic heart failure. They are considered disease-modifying agents and their use is strongly encouraged. Beta-blocker therapy must be initiated using low doses and only when patients are hemo-dynamically stable, with gradual dose titrations to prevent acute decompensation.

Evidence for amiodarone shows some reduction in sudden death, but these data are less compelling. Moreover, adverse effects and drug interactions complicate long-term amiodarone use. Use of class I (eg, flecainide, procainamide, propafenone) and other class III (sotalol) anti-arrhythmics to reduce sudden death is discouraged.

EVIDENCE-BASED ANSWER

Beta-blockers (class II antiarrhythmics) reduce sudden death and total mortality in patients with heart failure (strength of recommendation [SOR]: A, based on systematic reviews of randomized controlled trials). Amiodarone (class III) may reduce sudden death in heart failure (SOR: B, extrapolation from randomized controlled trials), but evidence is weak that it reduces total mortality, and it has significant side effects. Class I and other class III antiarrhythmic agents appear cause an increase in mortality due to sudden death in heart failure (SOR: B, extrapolations from randomized controlled trials).

 

Evidence summary

Antiarrhythmic agents have been studied in patients with heart failure because these persons have a high incidence of sudden death, presumably from ventricular arrhythmias. Although the implantable defibrillator is an alternative antiarrhythmic device that may be preferred for some patients, we restricted our review to pharmacologic antiarrhythmics.

The beta-blockers bisoprolol, carvedilol, and metoprolol1-3 were studied in large randomized controlled trials. The relative risk reduction (RRR) for sudden death ranged from 10% to 52% in the larger trials and 30% to 39% in meta-analyses.1-4 The absolute risk reduction (ARR) was about 2% to 3% per year for sudden death and 3% to 5% for total mortality (number needed to treat=20–33 per year).

These beta-blockers were well-tolerated, even in class IV New York Heart Association patients, and improved other endpoints. Although we cannot say whether the benefits are a class effect, they were seen with both beta-1 selective and nonselective agents.

Amiodarone was studied in 2 large randomized controlled trials enrolling patients with heart failure, in trials that included patients with or without heart failure at high risk for sudden death (usually post-myocardial infarction or with complex ventricular arrhythmias), and in meta-analyses.5-8 The largest randomized controlled trial in heart failure showed a significant ARR of 2.9% for sudden death,5 but was unblinded. The largest placebo-controlled trial in heart failure failed to detect a significant decrease in sudden death.6

Meta-analyses, weakened by heterogeneity and the inclusion of patients without heart failure, detected a significant 21% to 25% RRR for sudden death,7,8 and an ARR of 2% to 3% per year. The pooled data from the placebo-controlled heart failure trials showed nonsignificant trends: 1.6% per year ARR for sudden death, 0.6% per year for total mortality.

These possible benefits must be balanced against the risk of harm from amiodarone, including excess rates of pulmonary infiltrate (1.1% per year), thyroid dysfunction (6.8% per year), liver enzyme abnormalities (0.6% per year), neuropathy (0.3% per year), and bradycardia (1.6% per year), as well as a discontinuation rate of 41% compared with 27% for placebo.7 No evidence suggested that use of amiodarone in patients with heart failure increased mortality.

Class I antiarrhythmics and other class III agents have not been studied in heart failure trials, but were associated with increased mortality in studies of patients at high risk for ventricular arrhythmia,9,10 including patients with left ventricular dysfunction. Because this increase in mortality is thought to be due to proarrhythmic properties of the drugs, further trials in heart failure patients are unlikely to occur.

Recommendations from others

American College of Cardiology/American Heart Association (ACC/AHA),11 European Society of Cardiology (ESC),12 and Heart Failure Society of America (HFSA) guidelines13 address heart failure. ACC/AHA and ESC reports specifically mention that beta-blockers reduce sudden death. Both strongly support the use of beta-blockers in patients with heart failure.

ACC/AHA finds “conflicting evidence and/or a divergence of opinion about the usefulness/ efficacy” of amiodarone to prevent sudden death and advises: “routine use of amiodarone to prevent sudden death is not recommended.” The ESC and HFSA also recommend against routine use of amiodarone.

All 3 guidelines, however, state that for the control of symptomatic arrhythmias in heart failure, amiodarone is the antiarrhythmic agent of choice. All 3 also recommend not using class I or other class III agents in heart failure.

CLINICAL COMMENTARY

Beta-blockers reduce mortality in patients with heart failure
Joseph Saseen, PharmD, BCPS
University of Colorado Health Sciences Center, Denver

Numerous well-controlled clinical trials have conclusively demonstrated that beta-blockers reduce morbidity and mortality (including sudden death) in patients with systolic heart failure. They are considered disease-modifying agents and their use is strongly encouraged. Beta-blocker therapy must be initiated using low doses and only when patients are hemo-dynamically stable, with gradual dose titrations to prevent acute decompensation.

Evidence for amiodarone shows some reduction in sudden death, but these data are less compelling. Moreover, adverse effects and drug interactions complicate long-term amiodarone use. Use of class I (eg, flecainide, procainamide, propafenone) and other class III (sotalol) anti-arrhythmics to reduce sudden death is discouraged.

References

1. Effect of metoprolol CR/XL in chronic heart failure. Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet 1999;353:2001-2007.

2. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial Lancet 1999;353:9-13.

3. Packer M, Bristow MR, Cohn JN, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group. N Engl J Med 1996;334:1349-1355.

4. Lee S, Spencer A. Beta-blockers to reduce mortality in patients with systolic dysfunction: a meta-analysis. J Fam Pract 2001;50:499-504.

5. Doval HC, Nul DR, Grancelli HO, Perrone SV, Bortman GR, Curiel R. Randomised trial of low-dose amiodarone in severe congestive heart failure. Grupo de Estudio de la Sobrevida en la Insuficiencia Cardiaca en Argentina (GESICA). Lancet. 1994;344:493-498.

6. Singh SN, Fletcher RD, Fisher SG, et al. Amiodarone in patients with congestive heart failure and asymptomatic ventricular arrhythmia. Survival Trial of Antiarrhythmic Therapy in Congestive Heart Failure. N Engl J Med 1995;333:77-82.

7. Effect of prophylactic amiodarone on mortality after acute myocardial infarction and in congestive heart failure: meta-analysis of individual data from 6500 patients in randomised trials. Amiodarone Trials Meta-Analysis Investigators. Lancet 1997;350:1417-1424.

8. Piepoli M, Villani GO, Ponikowski P, Wright A, Flather MD, Coats AJ. Overview and meta-analysis of randomised trials of amiodarone in chronic heart failure. Int J Cardiol 1998;66:1-10.

9. Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. N Engl J Med 1991;324:781-788.

10. Connolly SJ. Meta-analysis of anti-arrhythmic drug trials. Am J Cardiol 1999;84:90R-93R.

11. Hunt SA, Baker DW, Chin MH, et al. ACC/AHA guidelines for the evaluation and management of chronic heart failure in the adult. Bethesda, Md: American College of Cardiology; 2001.

12. Priori SG, Aliot E, Blomstrom-Lundqvist C, et al. Task Force on Sudden Cardiac Death of the European Society of Cardiology. Eur Heart J 2001;22:1374-1450.

13. HFSA guidelines for management of patients with heart failure caused by left ventricular systolic dysfunction— pharmacological approaches. Heart Failure Society of merica. Pharmacotherapy 2000;20:495-522.

References

1. Effect of metoprolol CR/XL in chronic heart failure. Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet 1999;353:2001-2007.

2. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial Lancet 1999;353:9-13.

3. Packer M, Bristow MR, Cohn JN, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group. N Engl J Med 1996;334:1349-1355.

4. Lee S, Spencer A. Beta-blockers to reduce mortality in patients with systolic dysfunction: a meta-analysis. J Fam Pract 2001;50:499-504.

5. Doval HC, Nul DR, Grancelli HO, Perrone SV, Bortman GR, Curiel R. Randomised trial of low-dose amiodarone in severe congestive heart failure. Grupo de Estudio de la Sobrevida en la Insuficiencia Cardiaca en Argentina (GESICA). Lancet. 1994;344:493-498.

6. Singh SN, Fletcher RD, Fisher SG, et al. Amiodarone in patients with congestive heart failure and asymptomatic ventricular arrhythmia. Survival Trial of Antiarrhythmic Therapy in Congestive Heart Failure. N Engl J Med 1995;333:77-82.

7. Effect of prophylactic amiodarone on mortality after acute myocardial infarction and in congestive heart failure: meta-analysis of individual data from 6500 patients in randomised trials. Amiodarone Trials Meta-Analysis Investigators. Lancet 1997;350:1417-1424.

8. Piepoli M, Villani GO, Ponikowski P, Wright A, Flather MD, Coats AJ. Overview and meta-analysis of randomised trials of amiodarone in chronic heart failure. Int J Cardiol 1998;66:1-10.

9. Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. N Engl J Med 1991;324:781-788.

10. Connolly SJ. Meta-analysis of anti-arrhythmic drug trials. Am J Cardiol 1999;84:90R-93R.

11. Hunt SA, Baker DW, Chin MH, et al. ACC/AHA guidelines for the evaluation and management of chronic heart failure in the adult. Bethesda, Md: American College of Cardiology; 2001.

12. Priori SG, Aliot E, Blomstrom-Lundqvist C, et al. Task Force on Sudden Cardiac Death of the European Society of Cardiology. Eur Heart J 2001;22:1374-1450.

13. HFSA guidelines for management of patients with heart failure caused by left ventricular systolic dysfunction— pharmacological approaches. Heart Failure Society of merica. Pharmacotherapy 2000;20:495-522.

Issue
The Journal of Family Practice - 52(9)
Issue
The Journal of Family Practice - 52(9)
Page Number
714-724
Page Number
714-724
Publications
Publications
Topics
Article Type
Display Headline
Do antiarrhythmics prevent sudden death in patients with heart failure?
Display Headline
Do antiarrhythmics prevent sudden death in patients with heart failure?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

Do imaging studies aid diagnosis of acute sinusitis?

Article Type
Changed
Mon, 01/14/2019 - 10:58
Display Headline
Do imaging studies aid diagnosis of acute sinusitis?
EVIDENCE-BASED ANSWER

Accurate diagnosis of acute sinusitis in both children and adults depends on the history and clinical examination of the patient. While the clinical signs and symptoms of acute sinusitis are often difficult to distinguish from viral upper respiratory infection,1,2 such an assessment remains the best approach to diagnosing acute sinusitis (strength of recommendation [SOR]: A). There is no role for imaging in the diagnosis of acute sinusitis. For patients who have persistent symptoms, or those for whom surgery is being considered, some guidelines suggest that coronal computed tomography (CT) scan of the paranasal sinuses be considered (SOR: C, expert opinion).

 

Evidence summary

Three recent evidence-based guidelines3,4,5 suggest that children and adults with acute sinusitis may benefit from treatment with antibiotics more than those with rhinitis. Clinicians must develop a strategy for accurately diagnosing sinusitis to make sound treatment decisions. In the absence of a clear diagnosis of acute sinusitis, antibiotics are very unlikely to improve symptoms and are, therefore, not indicated.

Clinical evaluation. Berg1 studied 150 patients with clinical diagnoses of sinusitis and found that 85% of them had positive sinus puncture. In a review of the 11 studies that met evidence-based inclusion criteria, Varonen6 concluded that clinical evaluation has a sensitivity of roughly 0.75, whereas radiographic methodologies have sensitivities >0.80. In a prospective trial and subsequent review of the literature, Lindbaek7,8,9 suggests that several key clinical signs and symptoms can provide a level of sensitivity that approaches that of CT or magnetic resonance imaging (MRI), while enhancing specificity:

  • Purulent secretion reported as a symptom or found in the nasal cavity by the doctor
  • Pain in the teeth
  • Pain on bending forward (inconsistent findings between studies)
  • Two phases in the illness history
  • Elevated erythrocyte sedimentation rate or increased C-reactive protein
  • Symptoms for at least 7 days

Lau and colleagues5,10 reviewed 14 studies that compared various imaging studies with clinical evaluation or sinus puncture and aspiration with culture or both. A positive aspirate for bacterial pathogens was defined as the gold standard for diagnosis of sinusitis (Table).

X-ray vs sinus puncture. Depending on the criteria used to define a diagnosis of sinusitis on plain radiograph, estimates of sensitivity in these studies ranged from 0.41 to 0.90, and specificity estimates ranged from 0.61 to 0.85. Imaging studies that included “mucous membrane thickening” as a criterion for sinusitis were more sensitive but less specific than studies defining positive radiographs as “opacification of sinus.”

CT scan, MRI, ultrasound. While a CT scan is more sensitive than plain x-ray film,11 and MRI is more sensitive than a CT scan,12,13 the specificity of these studies is unclear. For example, in children and adults without symptoms of sinusitis, the prevalence of sinusitis signs on CT and MRI is 45% and 42%, respectively.6,7,14 In light of such findings, these imaging methodologies are better reserved for patients in whom surgery is being contemplated, or for whom chronic sinusitis is a concern. In the 1980s and 1990s, ultrasound was studied enthusiastically. Variability in test performance is great.6 Since the cost of this procedure is similar to that of a sinus CT, ultrasound is not indicated in the diagnostic evaluation of the sinuses.

Though the sensitivity and specificity of a clinical evaluation possibly could be enhanced with the use of imaging studies, diagnostic accuracy of acute disease is not sufficiently improved to justify the cost or inconvenience of such interventions.

TABLE
Sensitivity and specificity of imaging modalities in sinusitis

Diagnostic techniqueSensitivitySpecificity
X-rayVariableVariable
CT scanHighPoor
MRIHighPoor
Sinus punctureHighHigh
Clinical evaluationHighModerate

Recommendations from others

In a guideline on appropriate antibiotic use in sinusitis,4 endorsed by the Centers for Disease Control and Prevention, American Academy of Family Physicians, the American College of Physicians–American Society of Internal Medicine, and the Infectious Diseases Society of America, radiography is not recommended for the diagnosis of acute sinusitis. The guideline recommends that clinicians rely on duration of illness (at least 7 days) and severity of symptoms to make an accurate diagnosis of sinusitis.

The American Academy of Allergy, Asthma and Immunology15 guideline makes the following recommendations regarding imaging:

  • The use of imaging may be appropriate when there are vague symptoms, or poor response to initial management
  • Standard radiographs are insensitive, but may be used for diagnosis of acute sinus disease
  • CT is preferred for preoperative evaluation of the nose and paranasal sinuses
  • MRI is very sensitive for diagnosis of soft tissue disease in the frontal, maxillary, and sphenoid sinuses
  • Ultrasonography has limited utility but may be applicable in pregnant women and for determining the amount of retained secretions.
 

 

 

The Institute for Clinical Systems Improvement recommends that radiology be used only if initial treatment has failed, and notes that a primary goal of its guideline was to reduce the number of x-rays that physicians order for this diagnosis.16

The American College of Radiology’s criteria for sinusitis in the pediatric population ranked several radiographic studies based on their appropriateness for given clinical conditions. This review17 suggests that no imaging is appropriate if symptoms have persisted <10 days. For patients with symptoms lasting >10 days and with persistent fever, CT scan is recommended.

CLINICAL COMMENTARY

Jon Neher, MD
Valley Medical Center Family Practice Residency, Renton, Wash

In acute bacterial sinusitis, the history and physical have somewhat limited sensitivity and specificity. Unfortunately, imaging studies add little valuable information. Primary care physicians must therefore be reconciled to some degree of diagnostic error.

The risks associated with under-diagnosis are small, since most cases of mild sinusitis will resolve spontaneously without treatment. The risks of over-diagnosis include increased antibiotic costs, side effects, allergic reactions, and the development of resistant organisms. It is prudent, therefore, to make the diagnosis only when multiple suggestive historical and exam elements are present and to avoid giving antibiotics to patients with mild, nonspecific illnesses.

References

1. Berg O, Carenfelt C. Analysis of symptoms and clinical signs in the maxillary sinus empyema. Acta Otolaryngol 1988;105:343-349.

2. Williams JW, Jr, Simel DL, Roberts L, Samsa GP. Clinical evaluation for sinusitis. Making the diagnosis by history and physical examination. Ann Intern Med 1992;117:705-710.

3. Clinical practice guideline: management of sinusitis. Pediatrics 2001;108:798-808.

4. Snow V, Mottur-Pilson C, Hickner JM. Principles of appropriate antibiotic use for acute sinusitis in adults. Ann Intern Med 2001;134:495-497.

5. Lau J. Diagnosis and treatment of acute bacterial rhinosinusitis. Evidence Report/Technology Assessment No. 9. Rockville, MD: Agency for Health Care Policy and Research; 1999.

6. Varonen H, Makela M, Savolainen S, Laara E, Hilden J. Comparison of ultrasound, radiography, and clinical examination in the diagnosis of acute maxillary sinusitis: a systematic review. J Clin Epidemiol 2000;53:940-948.

7. Lindbaek M, Hjortdahl P. The clinical diagnosis of acute purulent sinusitis in general practice: a review. Br J Gen Pract 2002;52:491-495.

8. Lindbaek M, Hjortdahl P, Johnsen UL. Use of symptoms, signs, and blood tests to diagnose acute sinus infections in primary care: comparison with computed tomography. Fam Med 1996;28:183-188.

9. Lindbaek M, Johnsen UL, Kaastad E, et al. CT findings in general practice patients with suspected acute sinusitis. Acta Radiol 1996;37:708-713.

10. Benninger MS, Sedory Holzer SE, Lau J. Diagnosis and treatment of uncomplicated acute bacterial rhinosinusitis: summary of the Agency for Health Care Policy and Research evidence-based report. Otolaryngol Head Neck Surg 2000;122:1-7.

11. Cotter CS, Stringer S, Rust KR, Mancuso A. The role of computed tomography scans in evaluating sinus disease in pediatric patients. Int J Pediatr Otorhinolaryngol 1999;50:63-68.

12. Gordts F, Clement PA, Destryker A, Desprechins B, Kaufman L. Prevalence of sinusitis signs on MRI in a non-ENT paediatric population. Rhinology 1997;35:154-157.

13. Chong VF, Fan YF. Comparison of CT and MRI features in sinusitis. Eur J Radiol 1998;29:47-54.

14. Patel K, Chavda SV, Violaris N, Pahor AL. Incidental paranasal sinus inflammatory changes in a British population. J Laryngol Otol 1996;110:649-651.

15. Joint Task Force on Practice Parameters in Allergy, Asthma and Immunology : Joint Task Force summary statements on diagnosis and management of sinusitis.

16. Acute Sinusitis in Adults. Bloomington, Minn: Institute for Clinical Systems Improvement (ICSI), 2002. Available at: www.icsi.org. Accessed on June 17, 2003.

17. McAlister WH, Parker BR, Kushner DC, et al. Sinusitis in the pediatric population. American College of Radiology. ACR Appropriateness Criteria. Radiology 2000;215(Suppl):811-818.

Article PDF
Author and Disclosure Information

Jacob M. Reider, MD
Department of Family and Community Medicine, Albany Medical College, Albany, NY

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Issue
The Journal of Family Practice - 52(7)
Publications
Topics
Page Number
560-569
Sections
Author and Disclosure Information

Jacob M. Reider, MD
Department of Family and Community Medicine, Albany Medical College, Albany, NY

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Author and Disclosure Information

Jacob M. Reider, MD
Department of Family and Community Medicine, Albany Medical College, Albany, NY

Joan Nashelsky, MLS
Family Practice Inquiries Network, Iowa City, Iowa

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Accurate diagnosis of acute sinusitis in both children and adults depends on the history and clinical examination of the patient. While the clinical signs and symptoms of acute sinusitis are often difficult to distinguish from viral upper respiratory infection,1,2 such an assessment remains the best approach to diagnosing acute sinusitis (strength of recommendation [SOR]: A). There is no role for imaging in the diagnosis of acute sinusitis. For patients who have persistent symptoms, or those for whom surgery is being considered, some guidelines suggest that coronal computed tomography (CT) scan of the paranasal sinuses be considered (SOR: C, expert opinion).

 

Evidence summary

Three recent evidence-based guidelines3,4,5 suggest that children and adults with acute sinusitis may benefit from treatment with antibiotics more than those with rhinitis. Clinicians must develop a strategy for accurately diagnosing sinusitis to make sound treatment decisions. In the absence of a clear diagnosis of acute sinusitis, antibiotics are very unlikely to improve symptoms and are, therefore, not indicated.

Clinical evaluation. Berg1 studied 150 patients with clinical diagnoses of sinusitis and found that 85% of them had positive sinus puncture. In a review of the 11 studies that met evidence-based inclusion criteria, Varonen6 concluded that clinical evaluation has a sensitivity of roughly 0.75, whereas radiographic methodologies have sensitivities >0.80. In a prospective trial and subsequent review of the literature, Lindbaek7,8,9 suggests that several key clinical signs and symptoms can provide a level of sensitivity that approaches that of CT or magnetic resonance imaging (MRI), while enhancing specificity:

  • Purulent secretion reported as a symptom or found in the nasal cavity by the doctor
  • Pain in the teeth
  • Pain on bending forward (inconsistent findings between studies)
  • Two phases in the illness history
  • Elevated erythrocyte sedimentation rate or increased C-reactive protein
  • Symptoms for at least 7 days

Lau and colleagues5,10 reviewed 14 studies that compared various imaging studies with clinical evaluation or sinus puncture and aspiration with culture or both. A positive aspirate for bacterial pathogens was defined as the gold standard for diagnosis of sinusitis (Table).

X-ray vs sinus puncture. Depending on the criteria used to define a diagnosis of sinusitis on plain radiograph, estimates of sensitivity in these studies ranged from 0.41 to 0.90, and specificity estimates ranged from 0.61 to 0.85. Imaging studies that included “mucous membrane thickening” as a criterion for sinusitis were more sensitive but less specific than studies defining positive radiographs as “opacification of sinus.”

CT scan, MRI, ultrasound. While a CT scan is more sensitive than plain x-ray film,11 and MRI is more sensitive than a CT scan,12,13 the specificity of these studies is unclear. For example, in children and adults without symptoms of sinusitis, the prevalence of sinusitis signs on CT and MRI is 45% and 42%, respectively.6,7,14 In light of such findings, these imaging methodologies are better reserved for patients in whom surgery is being contemplated, or for whom chronic sinusitis is a concern. In the 1980s and 1990s, ultrasound was studied enthusiastically. Variability in test performance is great.6 Since the cost of this procedure is similar to that of a sinus CT, ultrasound is not indicated in the diagnostic evaluation of the sinuses.

Though the sensitivity and specificity of a clinical evaluation possibly could be enhanced with the use of imaging studies, diagnostic accuracy of acute disease is not sufficiently improved to justify the cost or inconvenience of such interventions.

TABLE
Sensitivity and specificity of imaging modalities in sinusitis

Diagnostic techniqueSensitivitySpecificity
X-rayVariableVariable
CT scanHighPoor
MRIHighPoor
Sinus punctureHighHigh
Clinical evaluationHighModerate

Recommendations from others

In a guideline on appropriate antibiotic use in sinusitis,4 endorsed by the Centers for Disease Control and Prevention, American Academy of Family Physicians, the American College of Physicians–American Society of Internal Medicine, and the Infectious Diseases Society of America, radiography is not recommended for the diagnosis of acute sinusitis. The guideline recommends that clinicians rely on duration of illness (at least 7 days) and severity of symptoms to make an accurate diagnosis of sinusitis.

The American Academy of Allergy, Asthma and Immunology15 guideline makes the following recommendations regarding imaging:

  • The use of imaging may be appropriate when there are vague symptoms, or poor response to initial management
  • Standard radiographs are insensitive, but may be used for diagnosis of acute sinus disease
  • CT is preferred for preoperative evaluation of the nose and paranasal sinuses
  • MRI is very sensitive for diagnosis of soft tissue disease in the frontal, maxillary, and sphenoid sinuses
  • Ultrasonography has limited utility but may be applicable in pregnant women and for determining the amount of retained secretions.
 

 

 

The Institute for Clinical Systems Improvement recommends that radiology be used only if initial treatment has failed, and notes that a primary goal of its guideline was to reduce the number of x-rays that physicians order for this diagnosis.16

The American College of Radiology’s criteria for sinusitis in the pediatric population ranked several radiographic studies based on their appropriateness for given clinical conditions. This review17 suggests that no imaging is appropriate if symptoms have persisted <10 days. For patients with symptoms lasting >10 days and with persistent fever, CT scan is recommended.

CLINICAL COMMENTARY

Jon Neher, MD
Valley Medical Center Family Practice Residency, Renton, Wash

In acute bacterial sinusitis, the history and physical have somewhat limited sensitivity and specificity. Unfortunately, imaging studies add little valuable information. Primary care physicians must therefore be reconciled to some degree of diagnostic error.

The risks associated with under-diagnosis are small, since most cases of mild sinusitis will resolve spontaneously without treatment. The risks of over-diagnosis include increased antibiotic costs, side effects, allergic reactions, and the development of resistant organisms. It is prudent, therefore, to make the diagnosis only when multiple suggestive historical and exam elements are present and to avoid giving antibiotics to patients with mild, nonspecific illnesses.

EVIDENCE-BASED ANSWER

Accurate diagnosis of acute sinusitis in both children and adults depends on the history and clinical examination of the patient. While the clinical signs and symptoms of acute sinusitis are often difficult to distinguish from viral upper respiratory infection,1,2 such an assessment remains the best approach to diagnosing acute sinusitis (strength of recommendation [SOR]: A). There is no role for imaging in the diagnosis of acute sinusitis. For patients who have persistent symptoms, or those for whom surgery is being considered, some guidelines suggest that coronal computed tomography (CT) scan of the paranasal sinuses be considered (SOR: C, expert opinion).

 

Evidence summary

Three recent evidence-based guidelines3,4,5 suggest that children and adults with acute sinusitis may benefit from treatment with antibiotics more than those with rhinitis. Clinicians must develop a strategy for accurately diagnosing sinusitis to make sound treatment decisions. In the absence of a clear diagnosis of acute sinusitis, antibiotics are very unlikely to improve symptoms and are, therefore, not indicated.

Clinical evaluation. Berg1 studied 150 patients with clinical diagnoses of sinusitis and found that 85% of them had positive sinus puncture. In a review of the 11 studies that met evidence-based inclusion criteria, Varonen6 concluded that clinical evaluation has a sensitivity of roughly 0.75, whereas radiographic methodologies have sensitivities >0.80. In a prospective trial and subsequent review of the literature, Lindbaek7,8,9 suggests that several key clinical signs and symptoms can provide a level of sensitivity that approaches that of CT or magnetic resonance imaging (MRI), while enhancing specificity:

  • Purulent secretion reported as a symptom or found in the nasal cavity by the doctor
  • Pain in the teeth
  • Pain on bending forward (inconsistent findings between studies)
  • Two phases in the illness history
  • Elevated erythrocyte sedimentation rate or increased C-reactive protein
  • Symptoms for at least 7 days

Lau and colleagues5,10 reviewed 14 studies that compared various imaging studies with clinical evaluation or sinus puncture and aspiration with culture or both. A positive aspirate for bacterial pathogens was defined as the gold standard for diagnosis of sinusitis (Table).

X-ray vs sinus puncture. Depending on the criteria used to define a diagnosis of sinusitis on plain radiograph, estimates of sensitivity in these studies ranged from 0.41 to 0.90, and specificity estimates ranged from 0.61 to 0.85. Imaging studies that included “mucous membrane thickening” as a criterion for sinusitis were more sensitive but less specific than studies defining positive radiographs as “opacification of sinus.”

CT scan, MRI, ultrasound. While a CT scan is more sensitive than plain x-ray film,11 and MRI is more sensitive than a CT scan,12,13 the specificity of these studies is unclear. For example, in children and adults without symptoms of sinusitis, the prevalence of sinusitis signs on CT and MRI is 45% and 42%, respectively.6,7,14 In light of such findings, these imaging methodologies are better reserved for patients in whom surgery is being contemplated, or for whom chronic sinusitis is a concern. In the 1980s and 1990s, ultrasound was studied enthusiastically. Variability in test performance is great.6 Since the cost of this procedure is similar to that of a sinus CT, ultrasound is not indicated in the diagnostic evaluation of the sinuses.

Though the sensitivity and specificity of a clinical evaluation possibly could be enhanced with the use of imaging studies, diagnostic accuracy of acute disease is not sufficiently improved to justify the cost or inconvenience of such interventions.

TABLE
Sensitivity and specificity of imaging modalities in sinusitis

Diagnostic techniqueSensitivitySpecificity
X-rayVariableVariable
CT scanHighPoor
MRIHighPoor
Sinus punctureHighHigh
Clinical evaluationHighModerate

Recommendations from others

In a guideline on appropriate antibiotic use in sinusitis,4 endorsed by the Centers for Disease Control and Prevention, American Academy of Family Physicians, the American College of Physicians–American Society of Internal Medicine, and the Infectious Diseases Society of America, radiography is not recommended for the diagnosis of acute sinusitis. The guideline recommends that clinicians rely on duration of illness (at least 7 days) and severity of symptoms to make an accurate diagnosis of sinusitis.

The American Academy of Allergy, Asthma and Immunology15 guideline makes the following recommendations regarding imaging:

  • The use of imaging may be appropriate when there are vague symptoms, or poor response to initial management
  • Standard radiographs are insensitive, but may be used for diagnosis of acute sinus disease
  • CT is preferred for preoperative evaluation of the nose and paranasal sinuses
  • MRI is very sensitive for diagnosis of soft tissue disease in the frontal, maxillary, and sphenoid sinuses
  • Ultrasonography has limited utility but may be applicable in pregnant women and for determining the amount of retained secretions.
 

 

 

The Institute for Clinical Systems Improvement recommends that radiology be used only if initial treatment has failed, and notes that a primary goal of its guideline was to reduce the number of x-rays that physicians order for this diagnosis.16

The American College of Radiology’s criteria for sinusitis in the pediatric population ranked several radiographic studies based on their appropriateness for given clinical conditions. This review17 suggests that no imaging is appropriate if symptoms have persisted <10 days. For patients with symptoms lasting >10 days and with persistent fever, CT scan is recommended.

CLINICAL COMMENTARY

Jon Neher, MD
Valley Medical Center Family Practice Residency, Renton, Wash

In acute bacterial sinusitis, the history and physical have somewhat limited sensitivity and specificity. Unfortunately, imaging studies add little valuable information. Primary care physicians must therefore be reconciled to some degree of diagnostic error.

The risks associated with under-diagnosis are small, since most cases of mild sinusitis will resolve spontaneously without treatment. The risks of over-diagnosis include increased antibiotic costs, side effects, allergic reactions, and the development of resistant organisms. It is prudent, therefore, to make the diagnosis only when multiple suggestive historical and exam elements are present and to avoid giving antibiotics to patients with mild, nonspecific illnesses.

References

1. Berg O, Carenfelt C. Analysis of symptoms and clinical signs in the maxillary sinus empyema. Acta Otolaryngol 1988;105:343-349.

2. Williams JW, Jr, Simel DL, Roberts L, Samsa GP. Clinical evaluation for sinusitis. Making the diagnosis by history and physical examination. Ann Intern Med 1992;117:705-710.

3. Clinical practice guideline: management of sinusitis. Pediatrics 2001;108:798-808.

4. Snow V, Mottur-Pilson C, Hickner JM. Principles of appropriate antibiotic use for acute sinusitis in adults. Ann Intern Med 2001;134:495-497.

5. Lau J. Diagnosis and treatment of acute bacterial rhinosinusitis. Evidence Report/Technology Assessment No. 9. Rockville, MD: Agency for Health Care Policy and Research; 1999.

6. Varonen H, Makela M, Savolainen S, Laara E, Hilden J. Comparison of ultrasound, radiography, and clinical examination in the diagnosis of acute maxillary sinusitis: a systematic review. J Clin Epidemiol 2000;53:940-948.

7. Lindbaek M, Hjortdahl P. The clinical diagnosis of acute purulent sinusitis in general practice: a review. Br J Gen Pract 2002;52:491-495.

8. Lindbaek M, Hjortdahl P, Johnsen UL. Use of symptoms, signs, and blood tests to diagnose acute sinus infections in primary care: comparison with computed tomography. Fam Med 1996;28:183-188.

9. Lindbaek M, Johnsen UL, Kaastad E, et al. CT findings in general practice patients with suspected acute sinusitis. Acta Radiol 1996;37:708-713.

10. Benninger MS, Sedory Holzer SE, Lau J. Diagnosis and treatment of uncomplicated acute bacterial rhinosinusitis: summary of the Agency for Health Care Policy and Research evidence-based report. Otolaryngol Head Neck Surg 2000;122:1-7.

11. Cotter CS, Stringer S, Rust KR, Mancuso A. The role of computed tomography scans in evaluating sinus disease in pediatric patients. Int J Pediatr Otorhinolaryngol 1999;50:63-68.

12. Gordts F, Clement PA, Destryker A, Desprechins B, Kaufman L. Prevalence of sinusitis signs on MRI in a non-ENT paediatric population. Rhinology 1997;35:154-157.

13. Chong VF, Fan YF. Comparison of CT and MRI features in sinusitis. Eur J Radiol 1998;29:47-54.

14. Patel K, Chavda SV, Violaris N, Pahor AL. Incidental paranasal sinus inflammatory changes in a British population. J Laryngol Otol 1996;110:649-651.

15. Joint Task Force on Practice Parameters in Allergy, Asthma and Immunology : Joint Task Force summary statements on diagnosis and management of sinusitis.

16. Acute Sinusitis in Adults. Bloomington, Minn: Institute for Clinical Systems Improvement (ICSI), 2002. Available at: www.icsi.org. Accessed on June 17, 2003.

17. McAlister WH, Parker BR, Kushner DC, et al. Sinusitis in the pediatric population. American College of Radiology. ACR Appropriateness Criteria. Radiology 2000;215(Suppl):811-818.

References

1. Berg O, Carenfelt C. Analysis of symptoms and clinical signs in the maxillary sinus empyema. Acta Otolaryngol 1988;105:343-349.

2. Williams JW, Jr, Simel DL, Roberts L, Samsa GP. Clinical evaluation for sinusitis. Making the diagnosis by history and physical examination. Ann Intern Med 1992;117:705-710.

3. Clinical practice guideline: management of sinusitis. Pediatrics 2001;108:798-808.

4. Snow V, Mottur-Pilson C, Hickner JM. Principles of appropriate antibiotic use for acute sinusitis in adults. Ann Intern Med 2001;134:495-497.

5. Lau J. Diagnosis and treatment of acute bacterial rhinosinusitis. Evidence Report/Technology Assessment No. 9. Rockville, MD: Agency for Health Care Policy and Research; 1999.

6. Varonen H, Makela M, Savolainen S, Laara E, Hilden J. Comparison of ultrasound, radiography, and clinical examination in the diagnosis of acute maxillary sinusitis: a systematic review. J Clin Epidemiol 2000;53:940-948.

7. Lindbaek M, Hjortdahl P. The clinical diagnosis of acute purulent sinusitis in general practice: a review. Br J Gen Pract 2002;52:491-495.

8. Lindbaek M, Hjortdahl P, Johnsen UL. Use of symptoms, signs, and blood tests to diagnose acute sinus infections in primary care: comparison with computed tomography. Fam Med 1996;28:183-188.

9. Lindbaek M, Johnsen UL, Kaastad E, et al. CT findings in general practice patients with suspected acute sinusitis. Acta Radiol 1996;37:708-713.

10. Benninger MS, Sedory Holzer SE, Lau J. Diagnosis and treatment of uncomplicated acute bacterial rhinosinusitis: summary of the Agency for Health Care Policy and Research evidence-based report. Otolaryngol Head Neck Surg 2000;122:1-7.

11. Cotter CS, Stringer S, Rust KR, Mancuso A. The role of computed tomography scans in evaluating sinus disease in pediatric patients. Int J Pediatr Otorhinolaryngol 1999;50:63-68.

12. Gordts F, Clement PA, Destryker A, Desprechins B, Kaufman L. Prevalence of sinusitis signs on MRI in a non-ENT paediatric population. Rhinology 1997;35:154-157.

13. Chong VF, Fan YF. Comparison of CT and MRI features in sinusitis. Eur J Radiol 1998;29:47-54.

14. Patel K, Chavda SV, Violaris N, Pahor AL. Incidental paranasal sinus inflammatory changes in a British population. J Laryngol Otol 1996;110:649-651.

15. Joint Task Force on Practice Parameters in Allergy, Asthma and Immunology : Joint Task Force summary statements on diagnosis and management of sinusitis.

16. Acute Sinusitis in Adults. Bloomington, Minn: Institute for Clinical Systems Improvement (ICSI), 2002. Available at: www.icsi.org. Accessed on June 17, 2003.

17. McAlister WH, Parker BR, Kushner DC, et al. Sinusitis in the pediatric population. American College of Radiology. ACR Appropriateness Criteria. Radiology 2000;215(Suppl):811-818.

Issue
The Journal of Family Practice - 52(7)
Issue
The Journal of Family Practice - 52(7)
Page Number
560-569
Page Number
560-569
Publications
Publications
Topics
Article Type
Display Headline
Do imaging studies aid diagnosis of acute sinusitis?
Display Headline
Do imaging studies aid diagnosis of acute sinusitis?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

What is the most effective nicotine replacement therapy?

Article Type
Changed
Mon, 01/14/2019 - 10:58
Display Headline
What is the most effective nicotine replacement therapy?
EVIDENCE-BASED ANSWER

No single nicotine replacement therapy is most effective for all smokers. All forms of nicotine replacement therapy (gum, transdermal patch, spray, inhaler, and lozenge) are equally effective, increasing smoking cessation rates by about 150% to 200%.1,2

A Cochrane Review found that 17% of smokers who had used nicotine replacement therapy successfully quit at follow-up vs 10% of smokers in the control group.1 Except in special circumstances (medical contraindications, smoking <10 cigarettes daily, pregnancy, or breastfeeding), all smokers attempting to quit should be offered nicotine replacement therapy (strength of recommendation [SOR]: A).3

Higher doses of nicotine gum or lozenge (4 mg vs 2 mg) increase quit rates in heavy smokers.1,2 Use of high-dose patches (>21 mg) may benefit heavy smokers or those relapsing due to nicotine withdrawal (SOR: B).3 For relapsed smokers, combination therapy improves long-term abstinence rates (estimated abstinence 28.6% vs 17.4% for monotherapy) (SOR: B).3

 

Evidence summary

A Cochrane Review of 110 trials evaluating the efficacy of nicotine replacement therapy in 35,600 smokers found higher quit rates among heavy smokers using 4-mg compared with 2-mg nicotine gum (odds ratio [OR], 2.67; 95% confidence interval [CI], 1.69–4.22).1 However, patients often chew too few pieces of nicotine gum daily, resulting in underdosing.3 Smokers should use the gum on a fixed schedule (at least 1 piece every 1 to 2 hours).3

The Cochrane Review finds borderline evidence of a small benefit in abstinence rates (OR, 1.21; 95% CI, 1.03–1.42) with higher-dose nicotine patches (>21 mg/24 hr or 15 mg/16 hr) for heavy or relapsed smokers.1 Combining methods that maintain constant drug levels (transdermal patch) with those having more rapid effects (gum, spray, inhaler, lozenge) is more effective than monotherapy (OR, 1.9; 95% CI, 1.3–2.6).3 Reserve combination therapy for smokers who relapse following monotherapy.

Regarding concerns about weight gain, all nicotine replacement therapies delay but do not prevent weight gain. There is a dose-response relationship between nicotine gum and weight gain: smokers who use more gum gain less weight.3 Although abstinence rates are comparable across the 5 available forms of nicotine replacement, smokers unwilling to give up oral and behavioral rituals of smoking may perceive the inhaler as being more helpful (Table 1).4

Decisions about the best form of therapy can be based on patient preference, on degree of nicotine dependence (a Fagerström Test of Nicotine Dependence Scale score ≥5 [Table 2], or habitually smoking the first cigarette within 30 minutes of awakening),5 or nicotine replacement therapy history, which includes number and outcome of previous quit attempts, specific method used, duration, side effects, and proper usage.

Recommendations from others

The Cochrane Review states: “All of the commercially available forms of nicotine replacement therapy are effective as part of a strategy to promote smoking cessation. They increase quit rates approximately 1.5 to 2 fold regardless of setting. Use of nicotine replacement therapy should be preferentially directed to smokers who are motivated to quit, and have high levels of nicotine dependency. Choice of which form to use should reflect patient needs, tolerability and cost considerations. Patches are likely to be easier to use than gum or nasal spray in primary care settings.”1

The US Department of Health and Human Services Clinical Practice Guideline states: “All patients attempting to quit should be encouraged to use effective pharmacotherapies for smoking cessation except in the presence of special circumstances.”3 Heavy smokers should use 4-mg nicotine gum. Combining the nicotine patch with a self-administered form of nicotine replacement therapy (gum or nicotine nasal spray) is more efficacious than a single form of therapy. Patients should be encouraged to use combined treatments if unable to quit using a single form of first-line pharmacotherapy.3

TABLE 1
Nicotine replacement therapy selection guide

 Moderate smokers (10–20 cigarettes/d)Heavy smokers (>30 cigarettes/d)Weight concerns
Gum (4 mg vs 2 mg gum enhances quit rates)(All nicotine replacement therapies delay weight gain, specifically nicotine gum)
Transdermal patch (Small benefit of dosing >21 mg) 
Inhaler   
Nasal spray   
Lozenge (Reserve 4 mg for heavy smokers) 
Combination   

TABLE 2
Fagerström test for level of nicotine dependence (abridged)

How soon after waking do you smoke first cigarette? ______ Points
 Less than 5 minutes: 3 points  
 5 to 30 minutes: 2 points  
 31 to 60 minutes: 1 point  
How many cigarettes do you smoke per day? ______ Points
 More than 30 per day: 3 points  
 21 to 30 per day: 2 points  
 11 to 20 per day: 1 point  
  ______ Total Points
Interpretation
 Total pointsLevel of dependenceNictotine replacement therapy
 5–6 pointsheavy nicotine dependenceconsider 21-mg nicotine patch
 3–4 pointsmoderate nicotine dependenceconsider 14-mg nicotine patch
 0–2 pointslight nicotine dependenceconsider 7-mg nicotine patch or no patch
CLINICAL COMMENTARY

Erik Lindbloom, MD, MSPH
Department of Family and Community Medicine, University of Missouri– Columbia

Now that nicotine replacement therapy is available over the counter, prescribers may not consider or discuss delivery options with patients as much as they did in the past. As this Clinical Inquiry illustrates, there are situations when one approach may be recommended over another.

For example, the relapsed smoker who has tried 1 nicotine replacement product may not even be aware that other methods, including combination therapy, are possible. Considering the enormous potential health improvement that is achieved through smoking cessation, this may be one of the most important topics to revisit regularly with patients.

References

1. Silagy C, Lancaster T, Stead L, Mant D, Fowler G. Nicotine replacement therapy for smoking cessation. The Cochrane Library, Volume (Issue 4), 2002.

2. Shiffman S, Dresler CM, Hajek P, Gilburt SJ, Targett DA, Strahs KR. Efficacy of a nicotine lozenge for smoking cessation. Arch Intern Med 2002;162:1267-1276.

3. Fiore MC, Bailey WC, Cohen SJ, et al. Treating Tobacco Use and Dependence. Clinical Practice Guideline. Rockville, Md: US Dept of Health and Human Services, Public Health Service, June 2000.

4. Schneider NG, Olmstead R, Nilsson F, Vaghaiwalla Mody F, Franzon M, Doan K. Efficacy of a nicotine inhaler in smoking cessation: a double-blind, placebo-controlled trial. Addiction 1996;91:1293-1306.

5. Heatherton TF, Kozlowski LT, Frecker RC, Fagerström KO. The Fagerström Test for Nicotine Dependence: a revision of the Fagerström Tolerance Questionnaire. Br J Addict 1991;86:1119-1127.

Article PDF
Author and Disclosure Information

Lisa J. Diefenbach, PsyD
Patrick O. Smith, PhD
Department of Family Medicine, University of Mississippi Medical Center, Jackson, Miss

Joan Nashelsky, MLS
Iowa City, Iowa

Issue
The Journal of Family Practice - 52(6)
Publications
Topics
Page Number
485-497
Sections
Author and Disclosure Information

Lisa J. Diefenbach, PsyD
Patrick O. Smith, PhD
Department of Family Medicine, University of Mississippi Medical Center, Jackson, Miss

Joan Nashelsky, MLS
Iowa City, Iowa

Author and Disclosure Information

Lisa J. Diefenbach, PsyD
Patrick O. Smith, PhD
Department of Family Medicine, University of Mississippi Medical Center, Jackson, Miss

Joan Nashelsky, MLS
Iowa City, Iowa

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

No single nicotine replacement therapy is most effective for all smokers. All forms of nicotine replacement therapy (gum, transdermal patch, spray, inhaler, and lozenge) are equally effective, increasing smoking cessation rates by about 150% to 200%.1,2

A Cochrane Review found that 17% of smokers who had used nicotine replacement therapy successfully quit at follow-up vs 10% of smokers in the control group.1 Except in special circumstances (medical contraindications, smoking <10 cigarettes daily, pregnancy, or breastfeeding), all smokers attempting to quit should be offered nicotine replacement therapy (strength of recommendation [SOR]: A).3

Higher doses of nicotine gum or lozenge (4 mg vs 2 mg) increase quit rates in heavy smokers.1,2 Use of high-dose patches (>21 mg) may benefit heavy smokers or those relapsing due to nicotine withdrawal (SOR: B).3 For relapsed smokers, combination therapy improves long-term abstinence rates (estimated abstinence 28.6% vs 17.4% for monotherapy) (SOR: B).3

 

Evidence summary

A Cochrane Review of 110 trials evaluating the efficacy of nicotine replacement therapy in 35,600 smokers found higher quit rates among heavy smokers using 4-mg compared with 2-mg nicotine gum (odds ratio [OR], 2.67; 95% confidence interval [CI], 1.69–4.22).1 However, patients often chew too few pieces of nicotine gum daily, resulting in underdosing.3 Smokers should use the gum on a fixed schedule (at least 1 piece every 1 to 2 hours).3

The Cochrane Review finds borderline evidence of a small benefit in abstinence rates (OR, 1.21; 95% CI, 1.03–1.42) with higher-dose nicotine patches (>21 mg/24 hr or 15 mg/16 hr) for heavy or relapsed smokers.1 Combining methods that maintain constant drug levels (transdermal patch) with those having more rapid effects (gum, spray, inhaler, lozenge) is more effective than monotherapy (OR, 1.9; 95% CI, 1.3–2.6).3 Reserve combination therapy for smokers who relapse following monotherapy.

Regarding concerns about weight gain, all nicotine replacement therapies delay but do not prevent weight gain. There is a dose-response relationship between nicotine gum and weight gain: smokers who use more gum gain less weight.3 Although abstinence rates are comparable across the 5 available forms of nicotine replacement, smokers unwilling to give up oral and behavioral rituals of smoking may perceive the inhaler as being more helpful (Table 1).4

Decisions about the best form of therapy can be based on patient preference, on degree of nicotine dependence (a Fagerström Test of Nicotine Dependence Scale score ≥5 [Table 2], or habitually smoking the first cigarette within 30 minutes of awakening),5 or nicotine replacement therapy history, which includes number and outcome of previous quit attempts, specific method used, duration, side effects, and proper usage.

Recommendations from others

The Cochrane Review states: “All of the commercially available forms of nicotine replacement therapy are effective as part of a strategy to promote smoking cessation. They increase quit rates approximately 1.5 to 2 fold regardless of setting. Use of nicotine replacement therapy should be preferentially directed to smokers who are motivated to quit, and have high levels of nicotine dependency. Choice of which form to use should reflect patient needs, tolerability and cost considerations. Patches are likely to be easier to use than gum or nasal spray in primary care settings.”1

The US Department of Health and Human Services Clinical Practice Guideline states: “All patients attempting to quit should be encouraged to use effective pharmacotherapies for smoking cessation except in the presence of special circumstances.”3 Heavy smokers should use 4-mg nicotine gum. Combining the nicotine patch with a self-administered form of nicotine replacement therapy (gum or nicotine nasal spray) is more efficacious than a single form of therapy. Patients should be encouraged to use combined treatments if unable to quit using a single form of first-line pharmacotherapy.3

TABLE 1
Nicotine replacement therapy selection guide

 Moderate smokers (10–20 cigarettes/d)Heavy smokers (>30 cigarettes/d)Weight concerns
Gum (4 mg vs 2 mg gum enhances quit rates)(All nicotine replacement therapies delay weight gain, specifically nicotine gum)
Transdermal patch (Small benefit of dosing >21 mg) 
Inhaler   
Nasal spray   
Lozenge (Reserve 4 mg for heavy smokers) 
Combination   

TABLE 2
Fagerström test for level of nicotine dependence (abridged)

How soon after waking do you smoke first cigarette? ______ Points
 Less than 5 minutes: 3 points  
 5 to 30 minutes: 2 points  
 31 to 60 minutes: 1 point  
How many cigarettes do you smoke per day? ______ Points
 More than 30 per day: 3 points  
 21 to 30 per day: 2 points  
 11 to 20 per day: 1 point  
  ______ Total Points
Interpretation
 Total pointsLevel of dependenceNictotine replacement therapy
 5–6 pointsheavy nicotine dependenceconsider 21-mg nicotine patch
 3–4 pointsmoderate nicotine dependenceconsider 14-mg nicotine patch
 0–2 pointslight nicotine dependenceconsider 7-mg nicotine patch or no patch
CLINICAL COMMENTARY

Erik Lindbloom, MD, MSPH
Department of Family and Community Medicine, University of Missouri– Columbia

Now that nicotine replacement therapy is available over the counter, prescribers may not consider or discuss delivery options with patients as much as they did in the past. As this Clinical Inquiry illustrates, there are situations when one approach may be recommended over another.

For example, the relapsed smoker who has tried 1 nicotine replacement product may not even be aware that other methods, including combination therapy, are possible. Considering the enormous potential health improvement that is achieved through smoking cessation, this may be one of the most important topics to revisit regularly with patients.

EVIDENCE-BASED ANSWER

No single nicotine replacement therapy is most effective for all smokers. All forms of nicotine replacement therapy (gum, transdermal patch, spray, inhaler, and lozenge) are equally effective, increasing smoking cessation rates by about 150% to 200%.1,2

A Cochrane Review found that 17% of smokers who had used nicotine replacement therapy successfully quit at follow-up vs 10% of smokers in the control group.1 Except in special circumstances (medical contraindications, smoking <10 cigarettes daily, pregnancy, or breastfeeding), all smokers attempting to quit should be offered nicotine replacement therapy (strength of recommendation [SOR]: A).3

Higher doses of nicotine gum or lozenge (4 mg vs 2 mg) increase quit rates in heavy smokers.1,2 Use of high-dose patches (>21 mg) may benefit heavy smokers or those relapsing due to nicotine withdrawal (SOR: B).3 For relapsed smokers, combination therapy improves long-term abstinence rates (estimated abstinence 28.6% vs 17.4% for monotherapy) (SOR: B).3

 

Evidence summary

A Cochrane Review of 110 trials evaluating the efficacy of nicotine replacement therapy in 35,600 smokers found higher quit rates among heavy smokers using 4-mg compared with 2-mg nicotine gum (odds ratio [OR], 2.67; 95% confidence interval [CI], 1.69–4.22).1 However, patients often chew too few pieces of nicotine gum daily, resulting in underdosing.3 Smokers should use the gum on a fixed schedule (at least 1 piece every 1 to 2 hours).3

The Cochrane Review finds borderline evidence of a small benefit in abstinence rates (OR, 1.21; 95% CI, 1.03–1.42) with higher-dose nicotine patches (>21 mg/24 hr or 15 mg/16 hr) for heavy or relapsed smokers.1 Combining methods that maintain constant drug levels (transdermal patch) with those having more rapid effects (gum, spray, inhaler, lozenge) is more effective than monotherapy (OR, 1.9; 95% CI, 1.3–2.6).3 Reserve combination therapy for smokers who relapse following monotherapy.

Regarding concerns about weight gain, all nicotine replacement therapies delay but do not prevent weight gain. There is a dose-response relationship between nicotine gum and weight gain: smokers who use more gum gain less weight.3 Although abstinence rates are comparable across the 5 available forms of nicotine replacement, smokers unwilling to give up oral and behavioral rituals of smoking may perceive the inhaler as being more helpful (Table 1).4

Decisions about the best form of therapy can be based on patient preference, on degree of nicotine dependence (a Fagerström Test of Nicotine Dependence Scale score ≥5 [Table 2], or habitually smoking the first cigarette within 30 minutes of awakening),5 or nicotine replacement therapy history, which includes number and outcome of previous quit attempts, specific method used, duration, side effects, and proper usage.

Recommendations from others

The Cochrane Review states: “All of the commercially available forms of nicotine replacement therapy are effective as part of a strategy to promote smoking cessation. They increase quit rates approximately 1.5 to 2 fold regardless of setting. Use of nicotine replacement therapy should be preferentially directed to smokers who are motivated to quit, and have high levels of nicotine dependency. Choice of which form to use should reflect patient needs, tolerability and cost considerations. Patches are likely to be easier to use than gum or nasal spray in primary care settings.”1

The US Department of Health and Human Services Clinical Practice Guideline states: “All patients attempting to quit should be encouraged to use effective pharmacotherapies for smoking cessation except in the presence of special circumstances.”3 Heavy smokers should use 4-mg nicotine gum. Combining the nicotine patch with a self-administered form of nicotine replacement therapy (gum or nicotine nasal spray) is more efficacious than a single form of therapy. Patients should be encouraged to use combined treatments if unable to quit using a single form of first-line pharmacotherapy.3

TABLE 1
Nicotine replacement therapy selection guide

 Moderate smokers (10–20 cigarettes/d)Heavy smokers (>30 cigarettes/d)Weight concerns
Gum (4 mg vs 2 mg gum enhances quit rates)(All nicotine replacement therapies delay weight gain, specifically nicotine gum)
Transdermal patch (Small benefit of dosing >21 mg) 
Inhaler   
Nasal spray   
Lozenge (Reserve 4 mg for heavy smokers) 
Combination   

TABLE 2
Fagerström test for level of nicotine dependence (abridged)

How soon after waking do you smoke first cigarette? ______ Points
 Less than 5 minutes: 3 points  
 5 to 30 minutes: 2 points  
 31 to 60 minutes: 1 point  
How many cigarettes do you smoke per day? ______ Points
 More than 30 per day: 3 points  
 21 to 30 per day: 2 points  
 11 to 20 per day: 1 point  
  ______ Total Points
Interpretation
 Total pointsLevel of dependenceNictotine replacement therapy
 5–6 pointsheavy nicotine dependenceconsider 21-mg nicotine patch
 3–4 pointsmoderate nicotine dependenceconsider 14-mg nicotine patch
 0–2 pointslight nicotine dependenceconsider 7-mg nicotine patch or no patch
CLINICAL COMMENTARY

Erik Lindbloom, MD, MSPH
Department of Family and Community Medicine, University of Missouri– Columbia

Now that nicotine replacement therapy is available over the counter, prescribers may not consider or discuss delivery options with patients as much as they did in the past. As this Clinical Inquiry illustrates, there are situations when one approach may be recommended over another.

For example, the relapsed smoker who has tried 1 nicotine replacement product may not even be aware that other methods, including combination therapy, are possible. Considering the enormous potential health improvement that is achieved through smoking cessation, this may be one of the most important topics to revisit regularly with patients.

References

1. Silagy C, Lancaster T, Stead L, Mant D, Fowler G. Nicotine replacement therapy for smoking cessation. The Cochrane Library, Volume (Issue 4), 2002.

2. Shiffman S, Dresler CM, Hajek P, Gilburt SJ, Targett DA, Strahs KR. Efficacy of a nicotine lozenge for smoking cessation. Arch Intern Med 2002;162:1267-1276.

3. Fiore MC, Bailey WC, Cohen SJ, et al. Treating Tobacco Use and Dependence. Clinical Practice Guideline. Rockville, Md: US Dept of Health and Human Services, Public Health Service, June 2000.

4. Schneider NG, Olmstead R, Nilsson F, Vaghaiwalla Mody F, Franzon M, Doan K. Efficacy of a nicotine inhaler in smoking cessation: a double-blind, placebo-controlled trial. Addiction 1996;91:1293-1306.

5. Heatherton TF, Kozlowski LT, Frecker RC, Fagerström KO. The Fagerström Test for Nicotine Dependence: a revision of the Fagerström Tolerance Questionnaire. Br J Addict 1991;86:1119-1127.

References

1. Silagy C, Lancaster T, Stead L, Mant D, Fowler G. Nicotine replacement therapy for smoking cessation. The Cochrane Library, Volume (Issue 4), 2002.

2. Shiffman S, Dresler CM, Hajek P, Gilburt SJ, Targett DA, Strahs KR. Efficacy of a nicotine lozenge for smoking cessation. Arch Intern Med 2002;162:1267-1276.

3. Fiore MC, Bailey WC, Cohen SJ, et al. Treating Tobacco Use and Dependence. Clinical Practice Guideline. Rockville, Md: US Dept of Health and Human Services, Public Health Service, June 2000.

4. Schneider NG, Olmstead R, Nilsson F, Vaghaiwalla Mody F, Franzon M, Doan K. Efficacy of a nicotine inhaler in smoking cessation: a double-blind, placebo-controlled trial. Addiction 1996;91:1293-1306.

5. Heatherton TF, Kozlowski LT, Frecker RC, Fagerström KO. The Fagerström Test for Nicotine Dependence: a revision of the Fagerström Tolerance Questionnaire. Br J Addict 1991;86:1119-1127.

Issue
The Journal of Family Practice - 52(6)
Issue
The Journal of Family Practice - 52(6)
Page Number
485-497
Page Number
485-497
Publications
Publications
Topics
Article Type
Display Headline
What is the most effective nicotine replacement therapy?
Display Headline
What is the most effective nicotine replacement therapy?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

What nonhormonal therapies are effective for postmenopausal vasomotor symptoms?

Article Type
Changed
Mon, 01/14/2019 - 10:58
Display Headline
What nonhormonal therapies are effective for postmenopausal vasomotor symptoms?
EVIDENCE-BASED ANSWER

Regular exercise may reduce vasomotor symptoms of menopause (strength of recommendation [SOR]: C—single observational study).1

Soy products/isoflavones, either through diet or supplementation, may reduce the incidence of hot flushes (SOR: D—inconsistent results of randomized trials).2

Clonidine, as an oral or transdermal preparation, reduces hot flushes (SOR: A—randomized clinical trials),3 as does gabapentin (SOR: A— single randomized clinical trial).4

In cancer patients who have had surgical menopause, selective serotonin reuptake inhibitors5 and megestrol6 (Megase) have been effective in reducing hot flushes (SOR: A; B for extrapolation to the general population).

Other therapies—including Bellergal (a combination of belladonna, ergotamine, and phenobarbital), methyldopa, evening primrose oil, mai quan, flaxseed, ginseng, and topical wild yam extract—have not been effective.7 Black cohosh may be effective, but the evidence for this is of poor quality (SOR: C). (See Table.)

TABLE
Nonhormonal therapies for postmenopausal vasomotor symptoms

AgentEffectiveSOR Comments
Soy/isoflavonesMaybeDMultiple RCTs with conflicting results, no formal meta-analysis. Does have a positive effect on lipid profile
Clonidine (Catapres)YesAMultiple small RCTs
Venlafaxine* (Effexor)YesBSingle RCT
Fluoxetine* (Prozac)YesBSingle RCT
Gabapentin (Neurontin)YesASingle RCT
Megestrol* (Megace)YesBSingle RCT
ExerciseMaybeCSingle observational study
Black cohoshMaybeCGerman E commission recommenda tion positive in 1989, but only 1 of 7 trials cited had placebo control. Recent RCT showed no benefit
Other: Bellergal, methyldopa, evening of effectNoCAll have been advocated but no positive trials for any evidence primrose oil, ginseng, wild yam extract, mai quan, flaxseed
*Trials conducted only with patients with breast cancer and interventional menopause, most of whom were on anti-estrogen therapy.
†See page 290 for a description of strength of recommendation.
SOR, strength of recommendation; RCT, randomized controlled trial
 

Evidence summary

Hormone replacement therapy (HRT) is the standard treatment for vasomotor symptoms of menopause, and it is effective for this indication. With recent studies showing no benefit from long-term HRT for menopausal women and increased adverse effects with its use (especially for women at risk for coronary heart disease), there has been increased interest in nonhormonal treatments for these symptoms.

A small number of randomized clinical trials have studied treatments other than HRT for the control of vasomotor symptoms of menopause. As a group, these trials have been short-term and have involved small numbers of patients. A disproportionate number of trials have been completed in breast cancer survivors, since these patients tend to have more severe vasomotor symptoms as a result of their anti-estrogenic therapies. Whether these results can be generalized to all postmenopausal women with vasomotor symptoms cannot be determined from the evidence.

Eleven randomized trials of soy protein/isoflavone used placebo controls. Results were mixed, with 7 trials showing no effect and 4 showing a reduction in hot flushes in comparison with placebo. Studies reporting a positive effect showed approximately a 15% reduction in episodes in comparison with placebo. In one 6-month trial, there was a correlation between hot flushes and urinary isoflavone excretion regardless of treatment group, suggesting a confounding effect of dietary intake of isoflavone.

Five of six randomized controlled trials of cloni-dine have shown a reduction in frequency of hot flushes ranging from 14%–50% compared with placebo. One trial, which used oral clonidine 0.1 mg daily, also reported an improved quality of life for the treatment group. A single randomized trial has shown that gabapentin, at a dose of 900 mg/day, is effective in reducing both frequency and severity of hot flashes.4

Trials of specific selective serotonin reuptake inhibitors have been completed in patients with vasomotor symptoms secondary to breast cancer therapies. Individual randomized controlled trials of venlafaxine and fluoxetine have proven these agents effective, and a preliminary open-labeled trial of paroxetine has also suggested benefit.

Several reviews suggest black cohosh may be effective for short-term treatment, and it is used in Germany for this indication. The trials we found were not placebo-controlled, however, and the safety of this agent is controversial. A single English-language placebo-controlled trial did not show any benefit for black cohosh.

 

 

 

Recommendations from others

The American College of Obstetrics and Gynecology clinical management guideline, “The use of botanicals for management of menopausal symptoms,” gives a level C recommendation (consensus and expert opinion) that “Soy and isoflavone may be helpful in short-term (2 years) treatment of vasomotor symptoms” and “black cohosh may be helpful in the short-term (6 months) treatment of women with vasomotor symptoms.” They note that “given the possibility that these compounds may interact with estrogen, these agents should not be considered free of potential harm in women with estrogen-dependent cancers.”8

The North American Menopause Society notes that behavior changes, such as moderate exercise and avoidance of hot-flush triggers, may prevent some hot flushes, although there is only anecdotal evidence for this. The efficacy of paced respiration—deep, slow abdominal breathing—to lessen hot flushes has been shown in a small trial. The society states that other alternative therapies have not been shown to be efficacious, except for moderate quantities of soy products.9 The Medical Letter says the evidence that phytoestrogens are helpful for menopausal women comes mostly from epidemiological studies. The long-term adverse effects of phytoestrogen consumption are not known.10

Clinical commentary

Laura B. Hansen, PharmD, BCPS
University of Colorado Health Sciences Center, Denver

Behavioral modifications may be the first approach to reduce the incidence of vasomotor symptoms in menopausal women. Recommendations include wearing several layers of breathable clothing; keeping a glass of cold water, ice pack, or small fan by the bedside and nearby at work; performing deep breathing relaxation techniques; and exercising routinely.

Effective nonhormonal treatments include phytoestrogens (2 years), black cohosh (6 months), clonidine, selective serotonin reuptake inhibitors, and venlafaxine. Overall, there are few well-designed clinical trials regarding the safety and effectiveness of botanical agents used for vasomotor symptoms. Since the Food and Drug Administration does not regulate the marketing and standardization of these products, patients should be advised to purchase products from reputable companies with internal standardization processes.

Additionally, patients should talk with their health care provider prior to initiating any alternative medication to avoid drug-disease and drug-drug interactions.

References

1. Ivarsson T, Spetz AC, Hammar M. Physical exercise and vasomotor symptoms in postmenopausal women. Maturitas 1998;29:139-146.

2. Upmalis DH, Lobo R, Bradley L, Warren M, Cone FL, Lamia CA. Vasomotor symptom relief by soy isoflavone extract tablets in postmenopausal women: a multicenter, double-blind, randomized, placebo-controlled study. Menopause 2000;7:236-242.

3. Pandya KJ, Raubertas RF, Flynn PJ, et al. Oral clonidine in postmenopausal patients with breast cancer experiencing tamoxifen-induced hot flashes: a University of Rochester Cancer Center Community Clinical Oncology Program study. Ann Intern Med 2000;132:788-793.

4. Guttuso T, Kurlan R, McDermott MP, et al. Gabapentin’s effects on hot flashes in postmenopausal women: a randomized controlled trial. Obstet Gynecol 2003;101:337-345.

5. Loprinzi CL, Michalak JC, Quella SK, et al. Megestrol acetate for the prevention of hot flashes. N Engl J Med 1994;331:347-352.

6. Loprinzi CL, Sloan JA, Perez EA, et al. Phase III evaluation of fluoxetine for treatment of hot flashes. J Clin Oncol 2002;20:1578-1583.

7. Taylor M. Alternative medicine and the perimenopause: an evidence-based review. Obstet Gynecol Clin North Am 2002;29:555-573.

8. Use of botanicals for management of menopausal symptoms. ACOG Pract Bull No. 28. Washington, DC: American College of Obstetricians and Gynecologists, 2001.

9. Clinical challenges of perimenopause: consensus opinions of the North American Menopause Society. Menopause 2000;7:5-13.

10. Phytoestrogens. Med Lett Drugs Ther 2000;1072:17-18.

Article PDF
Author and Disclosure Information

Dan Brewer, MD
Department of Family Medicine, University of Tennessee–Knoxville

Joan Nashelsky, MLS
Family Practice Inquiries Network, East Lansing, Mich

Issue
The Journal of Family Practice - 52(4)
Publications
Topics
Page Number
321-333
Sections
Author and Disclosure Information

Dan Brewer, MD
Department of Family Medicine, University of Tennessee–Knoxville

Joan Nashelsky, MLS
Family Practice Inquiries Network, East Lansing, Mich

Author and Disclosure Information

Dan Brewer, MD
Department of Family Medicine, University of Tennessee–Knoxville

Joan Nashelsky, MLS
Family Practice Inquiries Network, East Lansing, Mich

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Regular exercise may reduce vasomotor symptoms of menopause (strength of recommendation [SOR]: C—single observational study).1

Soy products/isoflavones, either through diet or supplementation, may reduce the incidence of hot flushes (SOR: D—inconsistent results of randomized trials).2

Clonidine, as an oral or transdermal preparation, reduces hot flushes (SOR: A—randomized clinical trials),3 as does gabapentin (SOR: A— single randomized clinical trial).4

In cancer patients who have had surgical menopause, selective serotonin reuptake inhibitors5 and megestrol6 (Megase) have been effective in reducing hot flushes (SOR: A; B for extrapolation to the general population).

Other therapies—including Bellergal (a combination of belladonna, ergotamine, and phenobarbital), methyldopa, evening primrose oil, mai quan, flaxseed, ginseng, and topical wild yam extract—have not been effective.7 Black cohosh may be effective, but the evidence for this is of poor quality (SOR: C). (See Table.)

TABLE
Nonhormonal therapies for postmenopausal vasomotor symptoms

AgentEffectiveSOR Comments
Soy/isoflavonesMaybeDMultiple RCTs with conflicting results, no formal meta-analysis. Does have a positive effect on lipid profile
Clonidine (Catapres)YesAMultiple small RCTs
Venlafaxine* (Effexor)YesBSingle RCT
Fluoxetine* (Prozac)YesBSingle RCT
Gabapentin (Neurontin)YesASingle RCT
Megestrol* (Megace)YesBSingle RCT
ExerciseMaybeCSingle observational study
Black cohoshMaybeCGerman E commission recommenda tion positive in 1989, but only 1 of 7 trials cited had placebo control. Recent RCT showed no benefit
Other: Bellergal, methyldopa, evening of effectNoCAll have been advocated but no positive trials for any evidence primrose oil, ginseng, wild yam extract, mai quan, flaxseed
*Trials conducted only with patients with breast cancer and interventional menopause, most of whom were on anti-estrogen therapy.
†See page 290 for a description of strength of recommendation.
SOR, strength of recommendation; RCT, randomized controlled trial
 

Evidence summary

Hormone replacement therapy (HRT) is the standard treatment for vasomotor symptoms of menopause, and it is effective for this indication. With recent studies showing no benefit from long-term HRT for menopausal women and increased adverse effects with its use (especially for women at risk for coronary heart disease), there has been increased interest in nonhormonal treatments for these symptoms.

A small number of randomized clinical trials have studied treatments other than HRT for the control of vasomotor symptoms of menopause. As a group, these trials have been short-term and have involved small numbers of patients. A disproportionate number of trials have been completed in breast cancer survivors, since these patients tend to have more severe vasomotor symptoms as a result of their anti-estrogenic therapies. Whether these results can be generalized to all postmenopausal women with vasomotor symptoms cannot be determined from the evidence.

Eleven randomized trials of soy protein/isoflavone used placebo controls. Results were mixed, with 7 trials showing no effect and 4 showing a reduction in hot flushes in comparison with placebo. Studies reporting a positive effect showed approximately a 15% reduction in episodes in comparison with placebo. In one 6-month trial, there was a correlation between hot flushes and urinary isoflavone excretion regardless of treatment group, suggesting a confounding effect of dietary intake of isoflavone.

Five of six randomized controlled trials of cloni-dine have shown a reduction in frequency of hot flushes ranging from 14%–50% compared with placebo. One trial, which used oral clonidine 0.1 mg daily, also reported an improved quality of life for the treatment group. A single randomized trial has shown that gabapentin, at a dose of 900 mg/day, is effective in reducing both frequency and severity of hot flashes.4

Trials of specific selective serotonin reuptake inhibitors have been completed in patients with vasomotor symptoms secondary to breast cancer therapies. Individual randomized controlled trials of venlafaxine and fluoxetine have proven these agents effective, and a preliminary open-labeled trial of paroxetine has also suggested benefit.

Several reviews suggest black cohosh may be effective for short-term treatment, and it is used in Germany for this indication. The trials we found were not placebo-controlled, however, and the safety of this agent is controversial. A single English-language placebo-controlled trial did not show any benefit for black cohosh.

 

 

 

Recommendations from others

The American College of Obstetrics and Gynecology clinical management guideline, “The use of botanicals for management of menopausal symptoms,” gives a level C recommendation (consensus and expert opinion) that “Soy and isoflavone may be helpful in short-term (2 years) treatment of vasomotor symptoms” and “black cohosh may be helpful in the short-term (6 months) treatment of women with vasomotor symptoms.” They note that “given the possibility that these compounds may interact with estrogen, these agents should not be considered free of potential harm in women with estrogen-dependent cancers.”8

The North American Menopause Society notes that behavior changes, such as moderate exercise and avoidance of hot-flush triggers, may prevent some hot flushes, although there is only anecdotal evidence for this. The efficacy of paced respiration—deep, slow abdominal breathing—to lessen hot flushes has been shown in a small trial. The society states that other alternative therapies have not been shown to be efficacious, except for moderate quantities of soy products.9 The Medical Letter says the evidence that phytoestrogens are helpful for menopausal women comes mostly from epidemiological studies. The long-term adverse effects of phytoestrogen consumption are not known.10

Clinical commentary

Laura B. Hansen, PharmD, BCPS
University of Colorado Health Sciences Center, Denver

Behavioral modifications may be the first approach to reduce the incidence of vasomotor symptoms in menopausal women. Recommendations include wearing several layers of breathable clothing; keeping a glass of cold water, ice pack, or small fan by the bedside and nearby at work; performing deep breathing relaxation techniques; and exercising routinely.

Effective nonhormonal treatments include phytoestrogens (2 years), black cohosh (6 months), clonidine, selective serotonin reuptake inhibitors, and venlafaxine. Overall, there are few well-designed clinical trials regarding the safety and effectiveness of botanical agents used for vasomotor symptoms. Since the Food and Drug Administration does not regulate the marketing and standardization of these products, patients should be advised to purchase products from reputable companies with internal standardization processes.

Additionally, patients should talk with their health care provider prior to initiating any alternative medication to avoid drug-disease and drug-drug interactions.

EVIDENCE-BASED ANSWER

Regular exercise may reduce vasomotor symptoms of menopause (strength of recommendation [SOR]: C—single observational study).1

Soy products/isoflavones, either through diet or supplementation, may reduce the incidence of hot flushes (SOR: D—inconsistent results of randomized trials).2

Clonidine, as an oral or transdermal preparation, reduces hot flushes (SOR: A—randomized clinical trials),3 as does gabapentin (SOR: A— single randomized clinical trial).4

In cancer patients who have had surgical menopause, selective serotonin reuptake inhibitors5 and megestrol6 (Megase) have been effective in reducing hot flushes (SOR: A; B for extrapolation to the general population).

Other therapies—including Bellergal (a combination of belladonna, ergotamine, and phenobarbital), methyldopa, evening primrose oil, mai quan, flaxseed, ginseng, and topical wild yam extract—have not been effective.7 Black cohosh may be effective, but the evidence for this is of poor quality (SOR: C). (See Table.)

TABLE
Nonhormonal therapies for postmenopausal vasomotor symptoms

AgentEffectiveSOR Comments
Soy/isoflavonesMaybeDMultiple RCTs with conflicting results, no formal meta-analysis. Does have a positive effect on lipid profile
Clonidine (Catapres)YesAMultiple small RCTs
Venlafaxine* (Effexor)YesBSingle RCT
Fluoxetine* (Prozac)YesBSingle RCT
Gabapentin (Neurontin)YesASingle RCT
Megestrol* (Megace)YesBSingle RCT
ExerciseMaybeCSingle observational study
Black cohoshMaybeCGerman E commission recommenda tion positive in 1989, but only 1 of 7 trials cited had placebo control. Recent RCT showed no benefit
Other: Bellergal, methyldopa, evening of effectNoCAll have been advocated but no positive trials for any evidence primrose oil, ginseng, wild yam extract, mai quan, flaxseed
*Trials conducted only with patients with breast cancer and interventional menopause, most of whom were on anti-estrogen therapy.
†See page 290 for a description of strength of recommendation.
SOR, strength of recommendation; RCT, randomized controlled trial
 

Evidence summary

Hormone replacement therapy (HRT) is the standard treatment for vasomotor symptoms of menopause, and it is effective for this indication. With recent studies showing no benefit from long-term HRT for menopausal women and increased adverse effects with its use (especially for women at risk for coronary heart disease), there has been increased interest in nonhormonal treatments for these symptoms.

A small number of randomized clinical trials have studied treatments other than HRT for the control of vasomotor symptoms of menopause. As a group, these trials have been short-term and have involved small numbers of patients. A disproportionate number of trials have been completed in breast cancer survivors, since these patients tend to have more severe vasomotor symptoms as a result of their anti-estrogenic therapies. Whether these results can be generalized to all postmenopausal women with vasomotor symptoms cannot be determined from the evidence.

Eleven randomized trials of soy protein/isoflavone used placebo controls. Results were mixed, with 7 trials showing no effect and 4 showing a reduction in hot flushes in comparison with placebo. Studies reporting a positive effect showed approximately a 15% reduction in episodes in comparison with placebo. In one 6-month trial, there was a correlation between hot flushes and urinary isoflavone excretion regardless of treatment group, suggesting a confounding effect of dietary intake of isoflavone.

Five of six randomized controlled trials of cloni-dine have shown a reduction in frequency of hot flushes ranging from 14%–50% compared with placebo. One trial, which used oral clonidine 0.1 mg daily, also reported an improved quality of life for the treatment group. A single randomized trial has shown that gabapentin, at a dose of 900 mg/day, is effective in reducing both frequency and severity of hot flashes.4

Trials of specific selective serotonin reuptake inhibitors have been completed in patients with vasomotor symptoms secondary to breast cancer therapies. Individual randomized controlled trials of venlafaxine and fluoxetine have proven these agents effective, and a preliminary open-labeled trial of paroxetine has also suggested benefit.

Several reviews suggest black cohosh may be effective for short-term treatment, and it is used in Germany for this indication. The trials we found were not placebo-controlled, however, and the safety of this agent is controversial. A single English-language placebo-controlled trial did not show any benefit for black cohosh.

 

 

 

Recommendations from others

The American College of Obstetrics and Gynecology clinical management guideline, “The use of botanicals for management of menopausal symptoms,” gives a level C recommendation (consensus and expert opinion) that “Soy and isoflavone may be helpful in short-term (2 years) treatment of vasomotor symptoms” and “black cohosh may be helpful in the short-term (6 months) treatment of women with vasomotor symptoms.” They note that “given the possibility that these compounds may interact with estrogen, these agents should not be considered free of potential harm in women with estrogen-dependent cancers.”8

The North American Menopause Society notes that behavior changes, such as moderate exercise and avoidance of hot-flush triggers, may prevent some hot flushes, although there is only anecdotal evidence for this. The efficacy of paced respiration—deep, slow abdominal breathing—to lessen hot flushes has been shown in a small trial. The society states that other alternative therapies have not been shown to be efficacious, except for moderate quantities of soy products.9 The Medical Letter says the evidence that phytoestrogens are helpful for menopausal women comes mostly from epidemiological studies. The long-term adverse effects of phytoestrogen consumption are not known.10

Clinical commentary

Laura B. Hansen, PharmD, BCPS
University of Colorado Health Sciences Center, Denver

Behavioral modifications may be the first approach to reduce the incidence of vasomotor symptoms in menopausal women. Recommendations include wearing several layers of breathable clothing; keeping a glass of cold water, ice pack, or small fan by the bedside and nearby at work; performing deep breathing relaxation techniques; and exercising routinely.

Effective nonhormonal treatments include phytoestrogens (2 years), black cohosh (6 months), clonidine, selective serotonin reuptake inhibitors, and venlafaxine. Overall, there are few well-designed clinical trials regarding the safety and effectiveness of botanical agents used for vasomotor symptoms. Since the Food and Drug Administration does not regulate the marketing and standardization of these products, patients should be advised to purchase products from reputable companies with internal standardization processes.

Additionally, patients should talk with their health care provider prior to initiating any alternative medication to avoid drug-disease and drug-drug interactions.

References

1. Ivarsson T, Spetz AC, Hammar M. Physical exercise and vasomotor symptoms in postmenopausal women. Maturitas 1998;29:139-146.

2. Upmalis DH, Lobo R, Bradley L, Warren M, Cone FL, Lamia CA. Vasomotor symptom relief by soy isoflavone extract tablets in postmenopausal women: a multicenter, double-blind, randomized, placebo-controlled study. Menopause 2000;7:236-242.

3. Pandya KJ, Raubertas RF, Flynn PJ, et al. Oral clonidine in postmenopausal patients with breast cancer experiencing tamoxifen-induced hot flashes: a University of Rochester Cancer Center Community Clinical Oncology Program study. Ann Intern Med 2000;132:788-793.

4. Guttuso T, Kurlan R, McDermott MP, et al. Gabapentin’s effects on hot flashes in postmenopausal women: a randomized controlled trial. Obstet Gynecol 2003;101:337-345.

5. Loprinzi CL, Michalak JC, Quella SK, et al. Megestrol acetate for the prevention of hot flashes. N Engl J Med 1994;331:347-352.

6. Loprinzi CL, Sloan JA, Perez EA, et al. Phase III evaluation of fluoxetine for treatment of hot flashes. J Clin Oncol 2002;20:1578-1583.

7. Taylor M. Alternative medicine and the perimenopause: an evidence-based review. Obstet Gynecol Clin North Am 2002;29:555-573.

8. Use of botanicals for management of menopausal symptoms. ACOG Pract Bull No. 28. Washington, DC: American College of Obstetricians and Gynecologists, 2001.

9. Clinical challenges of perimenopause: consensus opinions of the North American Menopause Society. Menopause 2000;7:5-13.

10. Phytoestrogens. Med Lett Drugs Ther 2000;1072:17-18.

References

1. Ivarsson T, Spetz AC, Hammar M. Physical exercise and vasomotor symptoms in postmenopausal women. Maturitas 1998;29:139-146.

2. Upmalis DH, Lobo R, Bradley L, Warren M, Cone FL, Lamia CA. Vasomotor symptom relief by soy isoflavone extract tablets in postmenopausal women: a multicenter, double-blind, randomized, placebo-controlled study. Menopause 2000;7:236-242.

3. Pandya KJ, Raubertas RF, Flynn PJ, et al. Oral clonidine in postmenopausal patients with breast cancer experiencing tamoxifen-induced hot flashes: a University of Rochester Cancer Center Community Clinical Oncology Program study. Ann Intern Med 2000;132:788-793.

4. Guttuso T, Kurlan R, McDermott MP, et al. Gabapentin’s effects on hot flashes in postmenopausal women: a randomized controlled trial. Obstet Gynecol 2003;101:337-345.

5. Loprinzi CL, Michalak JC, Quella SK, et al. Megestrol acetate for the prevention of hot flashes. N Engl J Med 1994;331:347-352.

6. Loprinzi CL, Sloan JA, Perez EA, et al. Phase III evaluation of fluoxetine for treatment of hot flashes. J Clin Oncol 2002;20:1578-1583.

7. Taylor M. Alternative medicine and the perimenopause: an evidence-based review. Obstet Gynecol Clin North Am 2002;29:555-573.

8. Use of botanicals for management of menopausal symptoms. ACOG Pract Bull No. 28. Washington, DC: American College of Obstetricians and Gynecologists, 2001.

9. Clinical challenges of perimenopause: consensus opinions of the North American Menopause Society. Menopause 2000;7:5-13.

10. Phytoestrogens. Med Lett Drugs Ther 2000;1072:17-18.

Issue
The Journal of Family Practice - 52(4)
Issue
The Journal of Family Practice - 52(4)
Page Number
321-333
Page Number
321-333
Publications
Publications
Topics
Article Type
Display Headline
What nonhormonal therapies are effective for postmenopausal vasomotor symptoms?
Display Headline
What nonhormonal therapies are effective for postmenopausal vasomotor symptoms?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

Does microalbuminuria screening in diabetes prevent complications?

Article Type
Changed
Tue, 05/03/2022 - 16:10
Display Headline
Does microalbuminuria screening in diabetes prevent complications?
EVIDENCE-BASED ANSWER

Screening diabetic patients for microalbuminuria identifies those who may benefit from treatments that delay progression to renal failure (strength of recommendation: B, based on extrapolation from Level 1 treatment studies of patients with microalbuminuria).

No research has determined the best method for screening for microalbuminuria, or whether screening in primary care populations will produce better long-term outcomes. No studies have examined the role of microalbuminuria screening after angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) have been instituted for other indications.

 

Evidence summary

Patients with diabetes mellitus have a 20% to 40% lifetime risk for development of nephropathy, and microalbuminuria is the earliest easily detectable marker of renal damage.1 Improved control of blood sugar2,3 and blood pressure4 decreases but does not completely prevent development of microalbuminuria and progression to overt kidney failure. ACE inhibitors and ARBs have been shown to diminish this progression even in the absence of hypertension (the latter in type 2 diabetes only) (Table).

No prospective randomized trials of screening have been reported. There is uncertainty about what method of screening is most effective and practical in primary care settings.10 Expert opinion recommends diagnosing microalbuminuria after 2 positive test results,1 but whether repeated tests improve diagnostic accuracy is still controversial.10

A large randomized controlled trial showing better long-term renal and vascular disease outcomes would be needed to give screening for microalbuminuria a strength of recommendation of A. Recruiting patients for such a study, and interpreting its results, would be difficult: many subjects would have other indications, such as hypertension or congestive heart failure, warranting use of potentially renoprotective medications.

TABLE
Reno- and cardioprotective efficacy of treatments for diabetic patients with microalbuminuria

DM typeMedicationNNTTime (years)To prevent endpoint
1ACE inhibitor (Captopril)7.9*2Clinical proteinuria5
2ACE inhibitor (Enalapril)6.3*5Macroalbuminuria6
2ACE inhibitor (Enalapril)2.4*7Significant proteinuria7
2ARB (Losartan)3.63.4End-stage renal disease8
2ACE inhibitor (Ramipril)44.5Cardiovascular disease9
*Normotensive subjects
†Myocardial infarction, revascularization procedure, stroke, cardiovascular death, congestive heart failure requiring hospitalization, overt nephropathy, renal dialysis, or laser treatment for retinopathy
DM, diabetes mellitus; NNT, number needed to treat; ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker

Recommendations from others

The American Diabetes Association recommends annual screening for microalbuminuria—after 5 years of established type 1 disease, and at time of diagnosis for type 2 diabetes without macroalbuminuria. Initial screening can use 1 of 3 methods: measurement of the albumin-to-creatinine ratio in a random, spot collection; 24-hour collection with creatinine, allowing the simultaneous measurement of creatinine clearance; timed (eg, 4-hour or overnight) collection. At least 2 of 3 tests measured within a 6-month period should show elevated levels before a patient is said to have microalbuminuria.1

CLINICAL COMMENTARY

Stephen A. Wilson, MD
University of Pittsburgh Medical Center, St. Margaret Family Practice Residency, Pittsburgh, Pa

Blood pressure control and ACE inhibition improve mortality and morbidity for patients with diabetes mellitus type 2. Therefore, maximize ACE inhibitor or ARB doses, as tolerated, and aim for a blood pressure of 110–120/70–80 mm Hg (130/85 mm Hg is the maximum).

Using this plan, I do not routinely screen for microalbuminuria—which is, at best, a surrogate marker for nephropathy and poor blood pressure control—unless I believe it will work as an educational and motivational tool for patients who are less committed to self-care.

If serum creatinine becomes elevated, a 24-hour urine collection to examine volume, creatinine clearance, and protein can be used to help develop a negotiated care plan with the patient, which may or may not include referral. Until there is different evidence about screening and treatment options for microalbuminuria, I see no need to screen when the above plan is in effect.

References

1. Standards of medical care for patients with diabetes mellitus. Diabetes Care 2002;25:S33-S49.

2. Effect of intensive therapy on the development and progression of diabetic nephropathy in the Diabetes Control and Complications Trial. The Diabetes Control and Complications Trial (DCCT) Research Group. Kidney Int 1995;47:1703-1720.

3. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study Group. Lancet 1998;352:837-853.

4. Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38 UK Prospective Diabetes Study Group. BMJ 1998;317:703-713.

5. Laffel LM, McGill JB, Gans DJ. The beneficial effect of angiotensin-converting enzyme inhibition with captopril on diabetic nephropathy in normotensive IDDM patients with microalbuminuria. North American Microalbuminuria Study Group. Am J Med 1995;99:497-504.

6. Ahmad J, Siddiqui MA, Ahmad H. Effective postponement of diabetic nephropathy with enalapril in normotensive type 2 diabetic patients with microalbuminuria. Diabetes Care 1997;20:1576-1581.

7. Ravid M, Lang R, Rachmani R, Lishner M. Long-term renoprotective effect of angiotensin-converting enzyme inhibition in non-insulin-dependent diabetes mellitus. A 7-year follow-up study. Arch Intern Med 1996;156:286-289.

8. Brenner BM, Cooper ME, de Zeeuw D, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 2001;345:861-869.

9. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE study. Heart Outcomes Prevention Evaluation (HOPE) Study Investigators. Lancet 2000;355:253-259.

10. Scheid DC, McCarthy LH, Lawler FH, Hamm RM, Reilly KEH. Screening for microalbuminuria to prevent nephropathy in patients with diabetes. A systematic review of the evidence. J Fam Pract 2001;50:661-s668.

Article PDF
Author and Disclosure Information

Wayne A. Hale, MD, MS
Moses Cone Health System Family Practice Residency, Greensboro, NC

Joan Nashelsky, MLS
Family Practice Inquiries Network, East Lansing, Mich

Issue
The Journal of Family Practice - 52(3)
Publications
Topics
Page Number
229-239
Sections
Author and Disclosure Information

Wayne A. Hale, MD, MS
Moses Cone Health System Family Practice Residency, Greensboro, NC

Joan Nashelsky, MLS
Family Practice Inquiries Network, East Lansing, Mich

Author and Disclosure Information

Wayne A. Hale, MD, MS
Moses Cone Health System Family Practice Residency, Greensboro, NC

Joan Nashelsky, MLS
Family Practice Inquiries Network, East Lansing, Mich

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Screening diabetic patients for microalbuminuria identifies those who may benefit from treatments that delay progression to renal failure (strength of recommendation: B, based on extrapolation from Level 1 treatment studies of patients with microalbuminuria).

No research has determined the best method for screening for microalbuminuria, or whether screening in primary care populations will produce better long-term outcomes. No studies have examined the role of microalbuminuria screening after angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) have been instituted for other indications.

 

Evidence summary

Patients with diabetes mellitus have a 20% to 40% lifetime risk for development of nephropathy, and microalbuminuria is the earliest easily detectable marker of renal damage.1 Improved control of blood sugar2,3 and blood pressure4 decreases but does not completely prevent development of microalbuminuria and progression to overt kidney failure. ACE inhibitors and ARBs have been shown to diminish this progression even in the absence of hypertension (the latter in type 2 diabetes only) (Table).

No prospective randomized trials of screening have been reported. There is uncertainty about what method of screening is most effective and practical in primary care settings.10 Expert opinion recommends diagnosing microalbuminuria after 2 positive test results,1 but whether repeated tests improve diagnostic accuracy is still controversial.10

A large randomized controlled trial showing better long-term renal and vascular disease outcomes would be needed to give screening for microalbuminuria a strength of recommendation of A. Recruiting patients for such a study, and interpreting its results, would be difficult: many subjects would have other indications, such as hypertension or congestive heart failure, warranting use of potentially renoprotective medications.

TABLE
Reno- and cardioprotective efficacy of treatments for diabetic patients with microalbuminuria

DM typeMedicationNNTTime (years)To prevent endpoint
1ACE inhibitor (Captopril)7.9*2Clinical proteinuria5
2ACE inhibitor (Enalapril)6.3*5Macroalbuminuria6
2ACE inhibitor (Enalapril)2.4*7Significant proteinuria7
2ARB (Losartan)3.63.4End-stage renal disease8
2ACE inhibitor (Ramipril)44.5Cardiovascular disease9
*Normotensive subjects
†Myocardial infarction, revascularization procedure, stroke, cardiovascular death, congestive heart failure requiring hospitalization, overt nephropathy, renal dialysis, or laser treatment for retinopathy
DM, diabetes mellitus; NNT, number needed to treat; ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker

Recommendations from others

The American Diabetes Association recommends annual screening for microalbuminuria—after 5 years of established type 1 disease, and at time of diagnosis for type 2 diabetes without macroalbuminuria. Initial screening can use 1 of 3 methods: measurement of the albumin-to-creatinine ratio in a random, spot collection; 24-hour collection with creatinine, allowing the simultaneous measurement of creatinine clearance; timed (eg, 4-hour or overnight) collection. At least 2 of 3 tests measured within a 6-month period should show elevated levels before a patient is said to have microalbuminuria.1

CLINICAL COMMENTARY

Stephen A. Wilson, MD
University of Pittsburgh Medical Center, St. Margaret Family Practice Residency, Pittsburgh, Pa

Blood pressure control and ACE inhibition improve mortality and morbidity for patients with diabetes mellitus type 2. Therefore, maximize ACE inhibitor or ARB doses, as tolerated, and aim for a blood pressure of 110–120/70–80 mm Hg (130/85 mm Hg is the maximum).

Using this plan, I do not routinely screen for microalbuminuria—which is, at best, a surrogate marker for nephropathy and poor blood pressure control—unless I believe it will work as an educational and motivational tool for patients who are less committed to self-care.

If serum creatinine becomes elevated, a 24-hour urine collection to examine volume, creatinine clearance, and protein can be used to help develop a negotiated care plan with the patient, which may or may not include referral. Until there is different evidence about screening and treatment options for microalbuminuria, I see no need to screen when the above plan is in effect.

EVIDENCE-BASED ANSWER

Screening diabetic patients for microalbuminuria identifies those who may benefit from treatments that delay progression to renal failure (strength of recommendation: B, based on extrapolation from Level 1 treatment studies of patients with microalbuminuria).

No research has determined the best method for screening for microalbuminuria, or whether screening in primary care populations will produce better long-term outcomes. No studies have examined the role of microalbuminuria screening after angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) have been instituted for other indications.

 

Evidence summary

Patients with diabetes mellitus have a 20% to 40% lifetime risk for development of nephropathy, and microalbuminuria is the earliest easily detectable marker of renal damage.1 Improved control of blood sugar2,3 and blood pressure4 decreases but does not completely prevent development of microalbuminuria and progression to overt kidney failure. ACE inhibitors and ARBs have been shown to diminish this progression even in the absence of hypertension (the latter in type 2 diabetes only) (Table).

No prospective randomized trials of screening have been reported. There is uncertainty about what method of screening is most effective and practical in primary care settings.10 Expert opinion recommends diagnosing microalbuminuria after 2 positive test results,1 but whether repeated tests improve diagnostic accuracy is still controversial.10

A large randomized controlled trial showing better long-term renal and vascular disease outcomes would be needed to give screening for microalbuminuria a strength of recommendation of A. Recruiting patients for such a study, and interpreting its results, would be difficult: many subjects would have other indications, such as hypertension or congestive heart failure, warranting use of potentially renoprotective medications.

TABLE
Reno- and cardioprotective efficacy of treatments for diabetic patients with microalbuminuria

DM typeMedicationNNTTime (years)To prevent endpoint
1ACE inhibitor (Captopril)7.9*2Clinical proteinuria5
2ACE inhibitor (Enalapril)6.3*5Macroalbuminuria6
2ACE inhibitor (Enalapril)2.4*7Significant proteinuria7
2ARB (Losartan)3.63.4End-stage renal disease8
2ACE inhibitor (Ramipril)44.5Cardiovascular disease9
*Normotensive subjects
†Myocardial infarction, revascularization procedure, stroke, cardiovascular death, congestive heart failure requiring hospitalization, overt nephropathy, renal dialysis, or laser treatment for retinopathy
DM, diabetes mellitus; NNT, number needed to treat; ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker

Recommendations from others

The American Diabetes Association recommends annual screening for microalbuminuria—after 5 years of established type 1 disease, and at time of diagnosis for type 2 diabetes without macroalbuminuria. Initial screening can use 1 of 3 methods: measurement of the albumin-to-creatinine ratio in a random, spot collection; 24-hour collection with creatinine, allowing the simultaneous measurement of creatinine clearance; timed (eg, 4-hour or overnight) collection. At least 2 of 3 tests measured within a 6-month period should show elevated levels before a patient is said to have microalbuminuria.1

CLINICAL COMMENTARY

Stephen A. Wilson, MD
University of Pittsburgh Medical Center, St. Margaret Family Practice Residency, Pittsburgh, Pa

Blood pressure control and ACE inhibition improve mortality and morbidity for patients with diabetes mellitus type 2. Therefore, maximize ACE inhibitor or ARB doses, as tolerated, and aim for a blood pressure of 110–120/70–80 mm Hg (130/85 mm Hg is the maximum).

Using this plan, I do not routinely screen for microalbuminuria—which is, at best, a surrogate marker for nephropathy and poor blood pressure control—unless I believe it will work as an educational and motivational tool for patients who are less committed to self-care.

If serum creatinine becomes elevated, a 24-hour urine collection to examine volume, creatinine clearance, and protein can be used to help develop a negotiated care plan with the patient, which may or may not include referral. Until there is different evidence about screening and treatment options for microalbuminuria, I see no need to screen when the above plan is in effect.

References

1. Standards of medical care for patients with diabetes mellitus. Diabetes Care 2002;25:S33-S49.

2. Effect of intensive therapy on the development and progression of diabetic nephropathy in the Diabetes Control and Complications Trial. The Diabetes Control and Complications Trial (DCCT) Research Group. Kidney Int 1995;47:1703-1720.

3. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study Group. Lancet 1998;352:837-853.

4. Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38 UK Prospective Diabetes Study Group. BMJ 1998;317:703-713.

5. Laffel LM, McGill JB, Gans DJ. The beneficial effect of angiotensin-converting enzyme inhibition with captopril on diabetic nephropathy in normotensive IDDM patients with microalbuminuria. North American Microalbuminuria Study Group. Am J Med 1995;99:497-504.

6. Ahmad J, Siddiqui MA, Ahmad H. Effective postponement of diabetic nephropathy with enalapril in normotensive type 2 diabetic patients with microalbuminuria. Diabetes Care 1997;20:1576-1581.

7. Ravid M, Lang R, Rachmani R, Lishner M. Long-term renoprotective effect of angiotensin-converting enzyme inhibition in non-insulin-dependent diabetes mellitus. A 7-year follow-up study. Arch Intern Med 1996;156:286-289.

8. Brenner BM, Cooper ME, de Zeeuw D, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 2001;345:861-869.

9. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE study. Heart Outcomes Prevention Evaluation (HOPE) Study Investigators. Lancet 2000;355:253-259.

10. Scheid DC, McCarthy LH, Lawler FH, Hamm RM, Reilly KEH. Screening for microalbuminuria to prevent nephropathy in patients with diabetes. A systematic review of the evidence. J Fam Pract 2001;50:661-s668.

References

1. Standards of medical care for patients with diabetes mellitus. Diabetes Care 2002;25:S33-S49.

2. Effect of intensive therapy on the development and progression of diabetic nephropathy in the Diabetes Control and Complications Trial. The Diabetes Control and Complications Trial (DCCT) Research Group. Kidney Int 1995;47:1703-1720.

3. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study Group. Lancet 1998;352:837-853.

4. Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38 UK Prospective Diabetes Study Group. BMJ 1998;317:703-713.

5. Laffel LM, McGill JB, Gans DJ. The beneficial effect of angiotensin-converting enzyme inhibition with captopril on diabetic nephropathy in normotensive IDDM patients with microalbuminuria. North American Microalbuminuria Study Group. Am J Med 1995;99:497-504.

6. Ahmad J, Siddiqui MA, Ahmad H. Effective postponement of diabetic nephropathy with enalapril in normotensive type 2 diabetic patients with microalbuminuria. Diabetes Care 1997;20:1576-1581.

7. Ravid M, Lang R, Rachmani R, Lishner M. Long-term renoprotective effect of angiotensin-converting enzyme inhibition in non-insulin-dependent diabetes mellitus. A 7-year follow-up study. Arch Intern Med 1996;156:286-289.

8. Brenner BM, Cooper ME, de Zeeuw D, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 2001;345:861-869.

9. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE study. Heart Outcomes Prevention Evaluation (HOPE) Study Investigators. Lancet 2000;355:253-259.

10. Scheid DC, McCarthy LH, Lawler FH, Hamm RM, Reilly KEH. Screening for microalbuminuria to prevent nephropathy in patients with diabetes. A systematic review of the evidence. J Fam Pract 2001;50:661-s668.

Issue
The Journal of Family Practice - 52(3)
Issue
The Journal of Family Practice - 52(3)
Page Number
229-239
Page Number
229-239
Publications
Publications
Topics
Article Type
Display Headline
Does microalbuminuria screening in diabetes prevent complications?
Display Headline
Does microalbuminuria screening in diabetes prevent complications?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Article PDF Media